1
|
Dwivedi A, Kumar A, Faruq M, Singh VK, Dwivedi N, Singh K, Hussain I, Parida S, Kumar Jha G, Kumar N, Joshi D. Co-occurrence of Parkinson's disease and Retinitis Pigmentosa: A genetic and in silico analysis. Neuroscience 2025; 565:519-526. [PMID: 39674535 DOI: 10.1016/j.neuroscience.2024.12.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 12/01/2024] [Accepted: 12/09/2024] [Indexed: 12/16/2024]
Abstract
INTRODUCTION Parkinson's disease (PD) is primarily driven by the protein Alpha Synuclein (A-Syn) accumulation. Synphilin-1 protein, encoded by the SNCAIP gene, which co-localizes with A-Syn is a known risk factor for PD. Retinitis pigmentosa (RP), is a cluster of retinal degenerative disorders, and Cyclic Nucleotide Gated channel subunit Alpha 1 (CNGA1) is one of the initial genes associated with RP. Patients with PD can have various kinds of visual dysfunction as a non-motor manifestation, but to date, CNGA1 mutation and RP as a PD associated visual symptom has not been reported. We report a mutation in the SNCAIP gene in a PD patient, not reported earlier, and its co-occurrence with RP-associated CNGA1 gene mutation. METHOD Whole exome sequencing (WES) of the patient DNA sample and in-silico protein-protein interaction (PPI) analysis performed to find out proteins interacting with SNCAIP relevant concerning reported mutation of SNCAIP and further, CNGA1 interaction with SNCAIP. RESULT We are reporting, a missense mutation (p.Thr64Ser) at the SNCAIP gene, co-occurring with a missense variation (p.Gly509Arg) in the CNGA1 gene. In silico PPI analysis suggests SIAH1 as an important protein affected by SNCAIP mutation. LGALS4 and SNCA (gene encoding A-Syn) are common interactors between SNCAIP and CNGA1. CONCLUSION The current study has determined the co-occurrence of RP and PD, whole exome sequencing ascertains the mutations in SNCAIP and CNGA1 genes, which could be the cause of PD and RP co-occurrence.
Collapse
Affiliation(s)
- Archana Dwivedi
- Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Anand Kumar
- Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Mohammed Faruq
- CSIR-Institute of Genomic and Integrative Biology, Mall Road, Delhi, India
| | - Varun Kumar Singh
- Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Nidhi Dwivedi
- Department of Community Medicine, NDMC Medical College and Hindu Rao Hospital, Delhi, India
| | - Kamaljeet Singh
- Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, AIIMS Delhi, India
| | - Ibrahim Hussain
- Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Swati Parida
- Kalinga Institute of Medical Sciences, Bhuvaneshwar, Odisha 751024, India
| | - Gaurab Kumar Jha
- CSIR-Institute of Genomic and Integrative Biology, Mall Road, Delhi, India
| | - Niraj Kumar
- Department of Neurology, AIIMS, Bibinagar, Telangana, India
| | - Deepika Joshi
- Institute of Medical Sciences, Banaras Hindu University, Varanasi, India.
| |
Collapse
|
2
|
Safreena N, Nair IC, Chandra G. Therapeutic potential of Parkin and its regulation in Parkinson's disease. Biochem Pharmacol 2024; 230:116600. [PMID: 39500382 DOI: 10.1016/j.bcp.2024.116600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/05/2024] [Accepted: 10/28/2024] [Indexed: 11/14/2024]
Abstract
Parkinson's disease (PD) is a debilitating neurodegenerative disorder characterized by the progressive loss of dopaminergic neurons in the midbrain substantia nigra, resulting in motor and non-motor symptoms. While the exact etiology of PD remains elusive, a growing body of evidence suggests that dysfunction in the parkin protein plays a pivotal role in the pathogenesis of the disease. Parkin is an E3 ubiquitin ligase that ubiquitinates substrate proteins to control a number of crucial cellular processes including protein catabolism, immune response, and cellular apoptosis.While autosomal recessive mutations in the PARK2 gene, which codes for parkin, are linked to an inherited form of early-onset PD, heterozygous mutations in PARK2 have also been reported in the more commonly occurring sporadic PD cases. Impairment of parkin's E3 ligase activity is believed to play a pathogenic role in both familial and sporadic forms of PD.This article provides an overview of the current understanding of the mechanistic basis of parkin's E3 ligase activity, its major physiological role in controlling cellular functions, and how these are disrupted in familial and sporadic PD. The second half of the manuscript explores the currently available and potential therapeutic strategies targeting parkin structure and/or function in order to slow down or mitigate the progressive neurodegeneration in PD.
Collapse
Affiliation(s)
- Narukkottil Safreena
- Cell Biology Laboratory, Center for Development and Aging Research, Inter University Center for Biomedical Research & Super Specialty Hospital, Mahatma Gandhi University Campus at Thalappady, Rubber Board PO, Kottayam 686009, Kerala, India
| | - Indu C Nair
- SAS SNDP Yogam College, Konni, Pathanamthitta 689691, Kerala, India
| | - Goutam Chandra
- Cell Biology Laboratory, Center for Development and Aging Research, Inter University Center for Biomedical Research & Super Specialty Hospital, Mahatma Gandhi University Campus at Thalappady, Rubber Board PO, Kottayam 686009, Kerala, India.
| |
Collapse
|
3
|
Ho HH, Wing SS. α-Synuclein ubiquitination - functions in proteostasis and development of Lewy bodies. Front Mol Neurosci 2024; 17:1498459. [PMID: 39600913 PMCID: PMC11588729 DOI: 10.3389/fnmol.2024.1498459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 10/22/2024] [Indexed: 11/29/2024] Open
Abstract
Synucleinopathies are neurodegenerative disorders characterized by the accumulation of α-synuclein containing Lewy bodies. Ubiquitination, a key post-translational modification, has been recognized as a pivotal regulator of α-synuclein's cellular dynamics, influencing its degradation, aggregation, and associated neurotoxicity. This review examines comprehensively the current understanding of α-synuclein ubiquitination and its role in the pathogenesis of synucleinopathies, particularly in the context of Parkinson's disease. We explore the molecular mechanisms responsible for α-synuclein ubiquitination, with a focus on the roles of E3 ligases and deubiquitinases implicated in the degradation process which occurs primarily through the endosomal lysosomal pathway. The review further discusses how the dysregulation of these mechanisms contributes to α-synuclein aggregation and LB formation and offers suggestions for future investigations into the role of α-synuclein ubiquitination. Understanding these processes may shed light on potential therapeutic avenues that can modulate α-synuclein ubiquitination to alleviate its pathological impact in synucleinopathies.
Collapse
Affiliation(s)
- Hung-Hsiang Ho
- Department of Medicine, McGill University and Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Simon S. Wing
- Department of Medicine, McGill University and Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| |
Collapse
|
4
|
Zhang F, Luo A, Liao S, Liu M, Zhang J, Xu Z. Progress of non‐motor symptoms in early‐onset Parkinson's disease. IBRAIN 2024. [DOI: 10.1002/ibra.12180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 09/08/2024] [Indexed: 01/03/2025]
Abstract
AbstractParkinson's disease (PD) is a common degenerative disease of the central nervous system that is characterized by movement disorders and non‐motor symptoms (NMSs). The associated NMSs primarily include neuropsychiatric symptoms, autonomic dysfunction, sleep‐wake disorders, pain, fatigue, and hyposmia. These NMSs can occur at any stage of PD, especially before the onset of motor symptoms, and may affect a patient's quality of life more than motor symptoms. Although PD is most commonly diagnosed in people over 65 years, some patients exhibit symptom onset before the age of 50, which is clinically known as early‐onset Parkinson's disease (EOPD). The high heterogeneity and incidence of EOPD‐associated NMSs can lead to the misdiagnosis of EOPD as other neurodegenerative diseases. In this review, we discuss the research progress related to NMSs in patients with EOPD, focusing on neuropsychiatric disorders, autonomic dysfunction, sleep disorders, and sensory impairment, and outline the association of NMSs with different genotypic alterations, with the aim of providing assistance in the clinical management of patients.
Collapse
Affiliation(s)
- Fanshi Zhang
- Department of Neurology Affiliated Hospital of Zunyi Medical University Zunyi China
| | - Aidi Luo
- Department of Neurology Affiliated Hospital of Zunyi Medical University Zunyi China
| | - Shusheng Liao
- Department of Neurology Affiliated Hospital of Zunyi Medical University Zunyi China
| | - Mei Liu
- Department of Neurology Affiliated Hospital of Zunyi Medical University Zunyi China
| | - Jun Zhang
- Department of Neurology Affiliated Hospital of Zunyi Medical University Zunyi China
| | - Zucai Xu
- Department of Neurology Affiliated Hospital of Zunyi Medical University Zunyi China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine Zunyi Medical University Zunyi China
| |
Collapse
|
5
|
Borgen M, Grill B. Ubiquitin ligase signalling networks shape presynaptic development, function and disease. J Physiol 2024:10.1113/JP286469. [PMID: 39360902 PMCID: PMC11965430 DOI: 10.1113/jp286469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 08/27/2024] [Indexed: 04/04/2025] Open
Abstract
Ubiquitin ligases are important regulators of nervous system development, function and disease. To date, numerous ubiquitin ligases have been discovered that regulate presynaptic biology. Here, we discuss recent findings on presynaptic ubiquitin ligases that include members from the three major ubiquitin ligase classes: RING, RBR and HECT. Several themes emerge based on findings across a range of model systems. A cadre of ubiquitin ligases is required presynaptically to orchestrate development and transmission at synapses. Multiple ubiquitin ligases deploy both enzymatic and non-enzymatic mechanisms, and act as hubs for signalling networks at the synapse. Both excitatory and inhibitory presynaptic terminals are influenced by ligase activity. Finally, there are several neurodevelopmental disorders and neurodegenerative diseases associated with presynaptic ubiquitin ligases. These findings highlight the growing prominence and biomedical relevance of the presynaptic ubiquitin ligase network.
Collapse
Affiliation(s)
- Melissa Borgen
- Department of Biomedical Engineering and Science, Florida Institute of Technology, Melbourne, USA
| | - Brock Grill
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, USA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, USA
- Department of Pharmacology, University of Washington School of Medicine, Seattle, USA
| |
Collapse
|
6
|
Bustillos BA, Cocker LT, Coban MA, Weber CA, Bredenberg JM, Boneski PK, Siuda J, Slawek J, Puschmann A, Narendra DP, Graff-Radford NR, Wszolek ZK, Dickson DW, Ross OA, Caulfield TR, Springer W, Fiesel FC. Structural and Functional Characterization of the Most Frequent Pathogenic PRKN Substitution p.R275W. Cells 2024; 13:1540. [PMID: 39329724 PMCID: PMC11430725 DOI: 10.3390/cells13181540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 09/28/2024] Open
Abstract
Mutations in the PINK1 and PRKN genes are the most frequent genetic cause of early-onset Parkinson disease. The pathogenic p.R275W substitution in PRKN is the most frequent substitution observed in patients, and thus far has been characterized mostly through overexpression models that suggest a possible gain of toxic misfunction. However, its effects under endogenous conditions are largely unknown. We used patient fibroblasts, isogenic neurons, and post-mortem human brain samples from carriers with and without PRKN p.R275W to assess functional impact. Immunoblot analysis and immunofluorescence were used to study mitophagy activation, and mitophagy execution was analyzed by flow cytometry of the reporter mitoKeima. The functional analysis was accompanied by structural investigation of PRKN p.R275W. We observed lower PRKN protein in fibroblasts with compound heterozygous p.R275W mutations. Isogenic neurons showed an allele-dose dependent decrease in PRKN protein. Lower PRKN protein levels were accompanied by diminished phosphorylated ubiquitin and decreased MFN2 modification. Mitochondrial degradation was also allele-dose dependently impaired. Consistently, PRKN protein levels were drastically reduced in human brain samples from p.R275W carriers. Finally, structural simulations showed significant changes in the closed form of PRKN p.R275W. Our data suggest that under endogenous conditions the p.R275W mutation results in a loss-of-function by destabilizing PRKN.
Collapse
Affiliation(s)
- Bernardo A. Bustillos
- Mayo Clinic, Department of Neuroscience, Jacksonville, FL 32224, USA; (B.A.B.); (L.T.C.); (M.A.C.); (C.A.W.); (J.M.B.); (D.W.D.); (O.A.R.); (T.R.C.)
| | - Liam T. Cocker
- Mayo Clinic, Department of Neuroscience, Jacksonville, FL 32224, USA; (B.A.B.); (L.T.C.); (M.A.C.); (C.A.W.); (J.M.B.); (D.W.D.); (O.A.R.); (T.R.C.)
| | - Mathew A. Coban
- Mayo Clinic, Department of Neuroscience, Jacksonville, FL 32224, USA; (B.A.B.); (L.T.C.); (M.A.C.); (C.A.W.); (J.M.B.); (D.W.D.); (O.A.R.); (T.R.C.)
| | - Caleb A. Weber
- Mayo Clinic, Department of Neuroscience, Jacksonville, FL 32224, USA; (B.A.B.); (L.T.C.); (M.A.C.); (C.A.W.); (J.M.B.); (D.W.D.); (O.A.R.); (T.R.C.)
| | - Jenny M. Bredenberg
- Mayo Clinic, Department of Neuroscience, Jacksonville, FL 32224, USA; (B.A.B.); (L.T.C.); (M.A.C.); (C.A.W.); (J.M.B.); (D.W.D.); (O.A.R.); (T.R.C.)
| | - Paige K. Boneski
- Mayo Clinic, Department of Neuroscience, Jacksonville, FL 32224, USA; (B.A.B.); (L.T.C.); (M.A.C.); (C.A.W.); (J.M.B.); (D.W.D.); (O.A.R.); (T.R.C.)
| | - Joanna Siuda
- Department of Neurology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland
| | - Jaroslaw Slawek
- Department of Neurology, St. Adalbert Hospital, 80-462 Gdansk, Poland;
- Division of Neurological and Psychiatric Nursing, Faculty of Health Sciences, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Andreas Puschmann
- Department of Clinical Sciences, Neurology, Lund University, 22100 Lund, Sweden;
- Department of Neurology, Skane University Hospital, 22185 Lund, Sweden
| | - Derek P. Narendra
- Inherited Movement Disorders Unit, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke (NINDS), NIH, Bethesda, MD 20892, USA;
| | | | - Zbigniew K. Wszolek
- Mayo Clinic, Graduate School of Biomedical, Sciences Neuroscience PhD Program, Jacksonville, FL 32224, USA;
| | - Dennis W. Dickson
- Mayo Clinic, Department of Neuroscience, Jacksonville, FL 32224, USA; (B.A.B.); (L.T.C.); (M.A.C.); (C.A.W.); (J.M.B.); (D.W.D.); (O.A.R.); (T.R.C.)
- Mayo Clinic, Graduate School of Biomedical, Sciences Neuroscience PhD Program, Jacksonville, FL 32224, USA;
| | - Owen A. Ross
- Mayo Clinic, Department of Neuroscience, Jacksonville, FL 32224, USA; (B.A.B.); (L.T.C.); (M.A.C.); (C.A.W.); (J.M.B.); (D.W.D.); (O.A.R.); (T.R.C.)
- Mayo Clinic, Graduate School of Biomedical, Sciences Neuroscience PhD Program, Jacksonville, FL 32224, USA;
| | - Thomas R. Caulfield
- Mayo Clinic, Department of Neuroscience, Jacksonville, FL 32224, USA; (B.A.B.); (L.T.C.); (M.A.C.); (C.A.W.); (J.M.B.); (D.W.D.); (O.A.R.); (T.R.C.)
- Mayo Clinic, Department of Neurosurgery, Jacksonville, FL 32224, USA
- Mayo Clinic, Department of Cancer Biology, Jacksonville, FL 32224, USA
- Mayo Clinic, Department of Biochemistry & Molecular Biology, Jacksonville, FL 32224, USA
- Mayo Clinic, Department of Computational Biology, Jacksonville, FL 32224, USA
| | - Wolfdieter Springer
- Mayo Clinic, Department of Neuroscience, Jacksonville, FL 32224, USA; (B.A.B.); (L.T.C.); (M.A.C.); (C.A.W.); (J.M.B.); (D.W.D.); (O.A.R.); (T.R.C.)
- Mayo Clinic, Graduate School of Biomedical, Sciences Neuroscience PhD Program, Jacksonville, FL 32224, USA;
| | - Fabienne C. Fiesel
- Mayo Clinic, Department of Neuroscience, Jacksonville, FL 32224, USA; (B.A.B.); (L.T.C.); (M.A.C.); (C.A.W.); (J.M.B.); (D.W.D.); (O.A.R.); (T.R.C.)
- Mayo Clinic, Graduate School of Biomedical, Sciences Neuroscience PhD Program, Jacksonville, FL 32224, USA;
| |
Collapse
|
7
|
Barbuti PA. A-Syn(ful) MAM: A Fresh Perspective on a Converging Domain in Parkinson's Disease. Int J Mol Sci 2024; 25:6525. [PMID: 38928232 PMCID: PMC11203789 DOI: 10.3390/ijms25126525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 06/03/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Parkinson's disease (PD) is a disease of an unknown origin. Despite that, decades of research have provided considerable evidence that alpha-synuclein (αSyn) is central to the pathogenesis of disease. Mitochondria-associated endoplasmic reticulum (ER) membranes (MAMs) are functional domains formed at contact sites between the ER and mitochondria, with a well-established function of MAMs being the control of lipid homeostasis within the cell. Additionally, there are numerous proteins localized or enriched at MAMs that have regulatory roles in several different molecular signaling pathways required for cellular homeostasis, such as autophagy and neuroinflammation. Alterations in several of these signaling pathways that are functionally associated with MAMs are found in PD. Taken together with studies that find αSyn localized at MAMs, this has implicated MAM (dys)function as a converging domain relevant to PD. This review will highlight the many functions of MAMs and provide an overview of the literature that finds αSyn, in addition to several other PD-related proteins, localized there. This review will also detail the direct interaction of αSyn and αSyn-interacting partners with specific MAM-resident proteins. In addition, recent studies exploring new methods to investigate MAMs will be discussed, along with some of the controversies regarding αSyn, including its several conformations and subcellular localizations. The goal of this review is to highlight and provide insight on a domain that is incompletely understood and, from a PD perspective, highlight those complex interactions that may hold the key to understanding the pathomechanisms underlying PD, which may lead to the targeted development of new therapeutic strategies.
Collapse
Affiliation(s)
- Peter A Barbuti
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
8
|
Kluge A, Borsche M, Streubel-Gallasch L, Gül T, Schaake S, Balck A, Prasuhn J, Campbell P, Morris HR, Schapira AH, Lohmann K, Brüggemann N, Rakovic A, Seibler P, Başak AN, Berg D, Klein C. α-Synuclein Pathology in PRKN-Linked Parkinson's Disease: New Insights from a Blood-Based Seed Amplification Assay. Ann Neurol 2024; 95:1173-1177. [PMID: 38546204 DOI: 10.1002/ana.26917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 03/04/2024] [Accepted: 03/06/2024] [Indexed: 05/18/2024]
Abstract
Pathogenic variants in PRKN cause early-onset Parkinson's disease (PD), while the role of alpha-synuclein in PRKN-PD remains uncertain. One study performed a blood-based alpha-synuclein seed amplification assay (SAA) in PRKN-PD, not detecting seed amplification in 17 PRKN-PD patients. By applying a methodologically different SAA focusing on neuron-derived extracellular vesicles, we demonstrated alpha-synuclein seed amplification in 8 of 13 PRKN-PD patients, challenging the view of PRKN-PD as a non-synucleinopathy. Moreover, we performed blinded replication of the neuron-derived extracellular vesicles-dependent SAA in idiopathic PD patients and healthy controls. In conclusion, blood-based neuron-derived extracellular vesicles-dependent SAA represents a promising biomarker to elucidate the underpinnings of (monogenic) PD. ANN NEUROL 2024;95:1173-1177.
Collapse
Affiliation(s)
- Annika Kluge
- Department of Neurology, University Hospital Schleswig-Holstein Campus Kiel and Kiel University, Kiel, Germany
| | - Max Borsche
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
- Department of Neurology, University Hospital Schleswig-Holstein Campus Lübeck and University of Lübeck, Lübeck, Germany
| | | | - Tuğçe Gül
- Neurodegeneration Research Laboratory (NDAL), Research Center for Translational Medicine (KUTTAM), University School of Medicine, Istanbul, Turkey
| | - Susen Schaake
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Alexander Balck
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
- Department of Neurology, University Hospital Schleswig-Holstein Campus Lübeck and University of Lübeck, Lübeck, Germany
| | - Jannik Prasuhn
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
- Department of Neurology, University Hospital Schleswig-Holstein Campus Lübeck and University of Lübeck, Lübeck, Germany
| | - Philip Campbell
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
- UCL Movement Disorders Center, University College London, London, UK
| | - Huw R Morris
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
- UCL Movement Disorders Center, University College London, London, UK
| | - Anthony H Schapira
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
- UCL Movement Disorders Center, University College London, London, UK
| | - Katja Lohmann
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Norbert Brüggemann
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
- Department of Neurology, University Hospital Schleswig-Holstein Campus Lübeck and University of Lübeck, Lübeck, Germany
| | | | - Philip Seibler
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - A Nazlı Başak
- Neurodegeneration Research Laboratory (NDAL), Research Center for Translational Medicine (KUTTAM), University School of Medicine, Istanbul, Turkey
| | - Daniela Berg
- Department of Neurology, University Hospital Schleswig-Holstein Campus Kiel and Kiel University, Kiel, Germany
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| |
Collapse
|
9
|
Zhao X, Yang C, Chen X, Sun Y, Liu W, Ge Q, Yang J. Characteristic fingerprint spectrum of α-synuclein mutants on terahertz time-domain spectroscopy. Biophys J 2024; 123:1264-1273. [PMID: 38615192 PMCID: PMC11140463 DOI: 10.1016/j.bpj.2024.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 01/02/2024] [Accepted: 04/11/2024] [Indexed: 04/15/2024] Open
Abstract
α-Synuclein, a presynaptic neuronal protein encoded by the SNCA gene, is involved in the pathogenesis of Parkinson's disease. Point mutations and multiplications of α-synuclein (A30P and A53T) are correlated with early-onset Parkinson's disease characterized by rapid progression and poor prognosis. Currently, the clinical identification of SNCA variants, especially disease-related A30P and A53T mutants, remains challenging and also time consuming. This study aimed to develop a novel label-free detection method for distinguishing the SNCA mutants using transmission terahertz (THz) time-domain spectroscopy. The protein was spin-coated onto the quartz to form a thin film, which was measured using THz time-domain spectroscopy. The spectral characteristics of THz broadband pulse waves of α-synuclein protein variants (SNCA wild type, A30P, and A53T) at different frequencies were analyzed via Fourier transform. The amplitude A intensity (AWT, AA30P, and AA53T) and peak occurrence time in THz time-domain spectroscopy sensitively distinguished the three protein variants. The phase φ difference in THz frequency domain followed the trend of φWT > φA30P > φA53T. There was a significant difference in THz frequency amplitude A' corresponding to the frequency ranging from 0.4 to 0.66 THz (A'A53T > A'A30P > A'WT). At a frequency of 0.4-0.6 THz, the transmission T of THz waves distinguished three variants (TA53T > TA30P > TWT), whereas there was no difference in the transmission T at 0.66 THz. The SNCA wild-type protein and two mutant variants (A30P and A53T) had distinct characteristic fingerprint spectra on THz time-domain spectroscopy. This novel label-free detection method has great potential for the differential diagnosis of Parkinson's disease subtypes.
Collapse
Affiliation(s)
- Xiaofang Zhao
- Department of Neurosurgery, Peking University Third Hospital, Beijing, China; Center for Precision Neurosurgery and Oncology of Peking University Health Science Center, Beijing, China
| | - Chenlong Yang
- Department of Neurosurgery, Peking University Third Hospital, Beijing, China; Center for Precision Neurosurgery and Oncology of Peking University Health Science Center, Beijing, China
| | - Xin Chen
- Department of Neurosurgery, Peking University Third Hospital, Beijing, China; Center for Precision Neurosurgery and Oncology of Peking University Health Science Center, Beijing, China
| | - Yu Sun
- Department of Neurosurgery, Peking University Third Hospital, Beijing, China; Center for Precision Neurosurgery and Oncology of Peking University Health Science Center, Beijing, China
| | - Weihai Liu
- Department of Neurosurgery, Peking University Third Hospital, Beijing, China; Center for Precision Neurosurgery and Oncology of Peking University Health Science Center, Beijing, China
| | - Qinggang Ge
- Department of Intensive Care Unit, Peking University Third Hospital, Beijing, China
| | - Jun Yang
- Department of Neurosurgery, Peking University Third Hospital, Beijing, China; Center for Precision Neurosurgery and Oncology of Peking University Health Science Center, Beijing, China.
| |
Collapse
|
10
|
Sharma K, Chib S, Gupta A, Singh R, Chalotra R. Interplay between α-synuclein and parkin genes: Insights of Parkinson's disease. Mol Biol Rep 2024; 51:586. [PMID: 38683365 DOI: 10.1007/s11033-024-09520-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 04/03/2024] [Indexed: 05/01/2024]
Abstract
Parkinson's disease (PD) is a complex and debilitating neurodegenerative disorder characterized by the progressive loss of dopaminergic neurons in the substantia nigra. The pathogenesis of PD is intimately linked to the roles of two key molecular players, α-synuclein (α-syn) and Parkin. Understanding the intricate interplay between α-syn and Parkin is essential for unravelling the molecular underpinnings of PD. Their roles in synaptic function and protein quality control underscore their significance in neuronal health. Dysregulation of these processes, as seen in PD, highlights the potential for targeted therapeutic strategies aimed at restoring normal protein homeostasis and mitigating neurodegeneration. Investigating the connections between α-syn, Parkin, and various pathological mechanisms provides insights into the complex web of factors contributing to PD pathogenesis and offers hope for the development of more effective treatments for this devastating neurological disorder. The present compilation provides an overview of their structures, regional and cellular locations, associations, physiological functions, and pathological roles in the context of PD.
Collapse
Affiliation(s)
- Kajal Sharma
- Department of Pharmacology, Central University of Punjab, Bathinda, 151401, India
| | - Shivani Chib
- Department of Pharmacology, Central University of Punjab, Bathinda, 151401, India
| | - Aniket Gupta
- Department of Pharmacology, Central University of Punjab, Bathinda, 151401, India
| | - Randhir Singh
- Department of Pharmacology, Central University of Punjab, Bathinda, 151401, India.
| | - Rishabh Chalotra
- Department of Pharmacology, Central University of Punjab, Bathinda, 151401, India
| |
Collapse
|
11
|
Fukuyama Y, Kubo M, Harada K. Neurotrophic Natural Products. PROGRESS IN THE CHEMISTRY OF ORGANIC NATURAL PRODUCTS 2024; 123:1-473. [PMID: 38340248 DOI: 10.1007/978-3-031-42422-9_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2024]
Abstract
Neurotrophins (NGF, BDNF, NT3, NT4) can decrease cell death, induce differentiation, as well as sustain the structure and function of neurons, which make them promising therapeutic agents for the treatment of neurodegenerative disorders. However, neurotrophins have not been very effective in clinical trials mostly because they cannot pass through the blood-brain barrier owing to being high-molecular-weight proteins. Thus, neurotrophin-mimic small molecules, which stimulate the synthesis of endogenous neurotrophins or enhance neurotrophic actions, may serve as promising alternatives to neurotrophins. Small-molecular-weight natural products, which have been used in dietary functional foods or in traditional medicines over the course of human history, have a great potential for the development of new therapeutic agents against neurodegenerative diseases such as Alzheimer's disease. In this contribution, a variety of natural products possessing neurotrophic properties such as neurogenesis, neurite outgrowth promotion (neuritogenesis), and neuroprotection are described, and a focus is made on the chemistry and biology of several neurotrophic natural products.
Collapse
Affiliation(s)
- Yoshiyasu Fukuyama
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, 770-8514, Japan.
| | - Miwa Kubo
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, 770-8514, Japan
| | - Kenichi Harada
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, 770-8514, Japan
| |
Collapse
|
12
|
Heß T, Themann P, Oehlwein C, Milani TL. Does Impaired Plantar Cutaneous Vibration Perception Contribute to Axial Motor Symptoms in Parkinson's Disease? Effects of Medication and Subthalamic Nucleus Deep Brain Stimulation. Brain Sci 2023; 13:1681. [PMID: 38137129 PMCID: PMC10742284 DOI: 10.3390/brainsci13121681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 11/28/2023] [Accepted: 12/03/2023] [Indexed: 12/24/2023] Open
Abstract
OBJECTIVE To investigate whether impaired plantar cutaneous vibration perception contributes to axial motor symptoms in Parkinson's disease (PD) and whether anti-parkinsonian medication and subthalamic nucleus deep brain stimulation (STN-DBS) show different effects. METHODS Three groups were evaluated: PD patients in the medication "on" state (PD-MED), PD patients in the medication "on" state and additionally "on" STN-DBS (PD-MED-DBS), as well as healthy subjects (HS) as reference. Motor performance was analyzed using a pressure distribution platform. Plantar cutaneous vibration perception thresholds (VPT) were investigated using a customized vibration exciter at 30 Hz. RESULTS Motor performance of PD-MED and PD-MED-DBS was characterized by greater postural sway, smaller limits of stability ranges, and slower gait due to shorter strides, fewer steps per minute, and broader stride widths compared to HS. Comparing patient groups, PD-MED-DBS showed better overall motor performance than PD-MED, particularly for the functional limits of stability and gait. VPTs were significantly higher for PD-MED compared to those of HS, which suggests impaired plantar cutaneous vibration perception in PD. However, PD-MED-DBS showed less impaired cutaneous vibration perception than PD-MED. CONCLUSIONS PD patients suffer from poor motor performance compared to healthy subjects. Anti-parkinsonian medication in tandem with STN-DBS seems to be superior for normalizing axial motor symptoms compared to medication alone. Plantar cutaneous vibration perception is impaired in PD patients, whereas anti-parkinsonian medication together with STN-DBS is superior for normalizing tactile cutaneous perception compared to medication alone. Consequently, based on our results and the findings of the literature, impaired plantar cutaneous vibration perception might contribute to axial motor symptoms in PD.
Collapse
Affiliation(s)
- Tobias Heß
- Department of Human Locomotion, Chemnitz University of Technology, 09126 Chemnitz, Germany
| | - Peter Themann
- Department of Neurology and Parkinson, Clinic at Tharandter Forest, 09633 Halsbruecke, Germany
| | - Christian Oehlwein
- Neurological Outpatient Clinic for Parkinson Disease and Deep Brain Stimulation, 07551 Gera, Germany
| | - Thomas L. Milani
- Department of Human Locomotion, Chemnitz University of Technology, 09126 Chemnitz, Germany
| |
Collapse
|
13
|
Liang Y, Zhong G, Ren M, Sun T, Li Y, Ye M, Ma C, Guo Y, Liu C. The Role of Ubiquitin-Proteasome System and Mitophagy in the Pathogenesis of Parkinson's Disease. Neuromolecular Med 2023; 25:471-488. [PMID: 37698835 DOI: 10.1007/s12017-023-08755-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 08/24/2023] [Indexed: 09/13/2023]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease that is mainly in middle-aged people and elderly people, and the pathogenesis of PD is complex and diverse. The ubiquitin-proteasome system (UPS) is a master regulator of neural development and the maintenance of brain structure and function. Dysfunction of components and substrates of this UPS has been linked to neurodegenerative diseases such as Parkinson's disease and Alzheimer's disease. Moreover, UPS can regulate α-synuclein misfolding and aggregation, mitophagy, neuroinflammation and oxidative stress to affect the development of PD. In the present study, we review the role of several related E3 ubiquitin ligases and deubiquitinating enzymes (DUBs) on the pathogenesis of PD such as Parkin, CHIP, USP8, etc. On this basis, we summarize the connections and differences of different E3 ubiquitin ligases in the pathogenesis, and elaborate on the regulatory progress of different DUBs on the pathogenesis of PD. Therefore, we can better understand their relationships and provide feasible and valuable therapeutic clues for UPS-related PD treatment research.
Collapse
Affiliation(s)
- Yu Liang
- School of Clinical Medicine, Bengbu Medical College, Bengbu, 233000, China
| | - Guangshang Zhong
- School of Life Sciences, Bengbu Medical College, Bengbu, 233000, China
| | - Mingxin Ren
- School of Clinical Medicine, Bengbu Medical College, Bengbu, 233000, China
| | - Tingting Sun
- School of Life Sciences, Bengbu Medical College, Bengbu, 233000, China
| | - Yangyang Li
- School of Life Sciences, Bengbu Medical College, Bengbu, 233000, China
| | - Ming Ye
- Department of Neurology, The First Affiliated Hospital of Bengbu Medical College, Bengbu Medical College, Bengbu, 233000, China
| | - Caiyun Ma
- School of Life Sciences, Bengbu Medical College, Bengbu, 233000, China
| | - Yu Guo
- School of Life Sciences, Bengbu Medical College, Bengbu, 233000, China.
| | - Changqing Liu
- School of Clinical Medicine, Bengbu Medical College, Bengbu, 233000, China.
- School of Life Sciences, Bengbu Medical College, Bengbu, 233000, China.
| |
Collapse
|
14
|
Mito S, Cheng B, Garcia BA, Yee Ooi X, Gonzalez D, Ruiz TC, Elisarraras FX, Tsin A. SAR study of niclosamide derivatives for neuroprotective function in SH-SY5Y neuroblastoma. Bioorg Med Chem Lett 2023; 96:129498. [PMID: 37804994 DOI: 10.1016/j.bmcl.2023.129498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 08/06/2023] [Accepted: 10/04/2023] [Indexed: 10/09/2023]
Abstract
Neurodegenerative disease is a debilitating and incurable condition that affects millions of people around the world. The loss of functions or malfunctions of neural cells are the causes of mortality. A proteosome inhibitor, MG132, is well known to cause neurodegeneration in vitro when model neuronal-derived cell lines are exposed to it. Niclosamide, an anthelmintic drug, which has been used to treat tapeworm infections for more than 50 years, has recently attracted renewed attention in drug repurposing because it has been found to be a good candidate in many drug development screenings. We recently found that all markers of MG132-induced neuronal cell toxicity, including the accumulation of ubiquitinated proteins, were prevented by the presence of niclosamide. In addition, niclosamide was shown to enhance autophagy induced by MG132. There results suggested that niclosamide could act as a neuroprotective agent. In the present study, niclosamide derivatives were synthesized, and the structure-activity relationship (SAR) were determined with respect to protein ubiquitination induced by MG132 and effect on cell survival signaling pathways for neuroprotective function. Our results indicate that phenol OH plays a significant role in neuroprotective activity while the niclosamide derivatives without Cl (5- or 2'-Cl) showed almost the same neuroprotective effect. 4'-NO2 can be replaced by N3 or CF3 whereas NH2 significantly decreased activity. These findings provide guidance for the development of new niclosamide analogues against neurodegenerative diseases including Parkinson's disease.
Collapse
Affiliation(s)
- Shizue Mito
- Department of Chemistry, The University of Texas Rio Grande Valley, Edinburg, TX 78539, USA; Department of Medical Education, School of Medicine, The University of Texas Rio Grande Valley, Edinburg 78541, USA.
| | - Benxu Cheng
- Department of Neuroscience, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Benjamin A Garcia
- Department of Chemistry, The University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
| | - Xin Yee Ooi
- Department of Neuroscience, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Daniela Gonzalez
- Department of Neuroscience, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Tess C Ruiz
- Department of Chemistry, The University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
| | - Francisco X Elisarraras
- Department of Neuroscience, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Andrew Tsin
- Department of Neuroscience, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| |
Collapse
|
15
|
Hou X, Chen TH, Koga S, Bredenberg JM, Faroqi AH, Delenclos M, Bu G, Wszolek ZK, Carr JA, Ross OA, McLean PJ, Murray ME, Dickson DW, Fiesel FC, Springer W. Alpha-synuclein-associated changes in PINK1-PRKN-mediated mitophagy are disease context dependent. Brain Pathol 2023; 33:e13175. [PMID: 37259617 PMCID: PMC10467041 DOI: 10.1111/bpa.13175] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 05/05/2023] [Indexed: 06/02/2023] Open
Abstract
Alpha-synuclein (αsyn) aggregates are pathological features of several neurodegenerative conditions including Parkinson disease (PD), dementia with Lewy bodies, and multiple system atrophy (MSA). Accumulating evidence suggests that mitochondrial dysfunction and impairments of the autophagic-lysosomal system can contribute to the deposition of αsyn, which in turn may interfere with health and function of these organelles in a potentially vicious cycle. Here we investigated a potential convergence of αsyn with the PINK1-PRKN-mediated mitochondrial autophagy pathway in cell models, αsyn transgenic mice, and human autopsy brain. PINK1 and PRKN identify and selectively label damaged mitochondria with phosphorylated ubiquitin (pS65-Ub) to mark them for degradation (mitophagy). We found that disease-causing multiplications of αsyn resulted in accumulation of the ubiquitin ligase PRKN in cells. This effect could be normalized by starvation-induced autophagy activation and by CRISPR/Cas9-mediated αsyn knockout. Upon acute mitochondrial damage, the increased levels of PRKN protein contributed to an enhanced pS65-Ub response. We further confirmed increased pS65-Ub-immunopositive signals in mouse brain with αsyn overexpression and in postmortem human disease brain. Of note, increased pS65-Ub was associated with neuronal Lewy body-type αsyn pathology, but not glial cytoplasmic inclusions of αsyn as seen in MSA. While our results add another layer of complexity to the crosstalk between αsyn and the PINK1-PRKN pathway, distinct mechanisms may underlie in cells and brain tissue despite similar outcomes. Notwithstanding, our finding suggests that pS65-Ub may be useful as a biomarker to discriminate different synucleinopathies and may serve as a potential therapeutic target for Lewy body disease.
Collapse
Affiliation(s)
- Xu Hou
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | | | - Shunsuke Koga
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | | | - Ayman H. Faroqi
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| | | | - Guojun Bu
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| | | | - Jonathan A. Carr
- Division of Neurology, Department of Medicine, Faculty of Medicine and Health SciencesStellenbosch UniversityCape TownSouth Africa
| | - Owen A. Ross
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| | - Pamela J. McLean
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| | - Melissa E. Murray
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| | - Dennis W. Dickson
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| | - Fabienne C. Fiesel
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| | - Wolfdieter Springer
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| |
Collapse
|
16
|
Chen X, Tsika E, Levine N, Moore DJ. VPS35 and α-Synuclein fail to interact to modulate neurodegeneration in rodent models of Parkinson's disease. Mol Neurodegener 2023; 18:51. [PMID: 37542299 PMCID: PMC10403858 DOI: 10.1186/s13024-023-00641-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 07/11/2023] [Indexed: 08/06/2023] Open
Abstract
BACKGROUND Mutations in the vacuolar protein sorting 35 ortholog (VPS35) gene cause late-onset, autosomal dominant Parkinson's disease (PD), with a single missense mutation (Asp620Asn, D620N) known to segregate with disease in families with PD. The VPS35 gene encodes a core component of the retromer complex, involved in the endosomal sorting and recycling of transmembrane cargo proteins. VPS35-linked PD is clinically indistinguishable from sporadic PD, although it is not yet known whether VPS35-PD brains exhibit α-synuclein-positive brainstem Lewy pathology that is characteristic of sporadic cases. Prior studies have suggested a functional interaction between VPS35 and the PD-linked gene product α-synuclein in lower organisms, where VPS35 deletion enhances α-synuclein-induced toxicity. In mice, VPS35 overexpression is reported to rescue hippocampal neuronal loss in human α-synuclein transgenic mice, potentially suggesting a retromer deficiency in these mice. METHODS Here, we employ multiple well-established genetic rodent models to explore a functional or pathological interaction between VPS35 and α-synuclein in vivo. RESULTS We find that endogenous α-synuclein is dispensable for nigrostriatal pathway dopaminergic neurodegeneration induced by the viral-mediated delivery of human D620N VPS35 in mice, suggesting that α-synuclein does not operate downstream of VPS35. We next evaluated retromer levels in affected brain regions from human A53T-α-synuclein transgenic mice, but find normal levels of the core subunits VPS35, VPS26 or VPS29. We further find that heterozygous VPS35 deletion fails to alter the lethal neurodegenerative phenotype of these A53T-α-synuclein transgenic mice, suggesting the absence of retromer deficiency in this PD model. Finally, we explored the neuroprotective capacity of increasing VPS35 expression in a viral-based human wild-type α-synuclein rat model of PD. However, we find that the overexpression of wild-type VPS35 is not sufficient for protection against α-synuclein-induced nigral dopaminergic neurodegeneration, α-synuclein pathology and reactive gliosis. CONCLUSION Collectively, our data suggest a limited interaction of VPS35 and α-synuclein in neurodegenerative models of PD, and do not provide support for their interaction within a common pathophysiological pathway.
Collapse
Affiliation(s)
- Xi Chen
- Department of Neurodegenerative Science, Van Andel Institute, 333 Bostwick Ave NE, Grand Rapids, MI, 49503, USA
| | - Elpida Tsika
- Brain Mind Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Vaud, 1015, Switzerland
- AC Immune SA, EPFL Innovation Park, Lausanne, 1015, Switzerland
| | - Nathan Levine
- Department of Neurodegenerative Science, Van Andel Institute, 333 Bostwick Ave NE, Grand Rapids, MI, 49503, USA
| | - Darren J Moore
- Department of Neurodegenerative Science, Van Andel Institute, 333 Bostwick Ave NE, Grand Rapids, MI, 49503, USA.
| |
Collapse
|
17
|
Yang Y, Zhang T, Liu L, Shan L, Hu X, Yang L, Gao F, Xiaoli W, Li H. Efficacy and safety of botulinum toxin for treating motor dysfunction in patients with Parkinson's disease: a systematic review and meta-analysis. BMJ Open 2023; 13:e060274. [PMID: 37328181 PMCID: PMC10277107 DOI: 10.1136/bmjopen-2021-060274] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 05/26/2023] [Indexed: 06/18/2023] Open
Abstract
OBJECTIVE To evaluate the efficacy and safety of botulinum toxin (BTX) for motor dysfunction in Parkinson's disease (PD). DESIGN Systematic review and meta-analysis. DATA SOURCES Searches of PubMed, EMBASE and the Cochrane Library, from database inception to 20 October 2022. ELIGIBILITY CRITERIA Studies reported in English with adult PD patients treated with BTX. DATA EXTRACTION AND SYNTHESIS Primary outcomes were United Parkinson's Disease Rate Scale Section (UPDRS) III (or its items) and Visual Analogue Scale (VAS). Secondary outcomes were UPDRS-II (or its items), Freezing of Gait Questionnaire (FOG-Q), Timed Up and Go test (TUG) and treatment-related adverse events (TRAEs). Mean difference (MD) or standardised MD (SMD) before and after treatment with 95% CIs were used for continuous variables and risk ratios (RRs) with 95% CIs was used for TRAEs. RESULTS Six randomised controlled trials (RCTs) and six non-RCTs (case series) were included (ntotal=224 participants, nRCT=165). No significant difference was found in pooled results of UPDRS-III (available in four RCTs and two non-RCTs, SMD=-0.19, 95% CI -0.98 to 0.60), UPDRS-II (four RCTs and one non-RCT, SMD=-0.55, 95% CI -1.22 to 0.13), FOG-Q (one RCT and one non-RCT, SMD=0.53, 95% CI -1.93 to 2.98) or the risk of TRAEs (five RCTs, RR 0.87, 95% CI 0.37 to 2.01). Significant decreases were found in pooled VAS score (three RCTs and five non-RCTs, MD=-2.14, 95% CI -3.05 to -1.23) and TUG (MD=-2.06, 95% CI -2.91 to -1.20) after BTX treatment. CONCLUSIONS BTX may not be associated with motor symptoms alleviation, although it benefits pain alleviation and functional mobility improvement.
Collapse
Affiliation(s)
- Yuqi Yang
- Department of Neurorehabilitation, China Rehabilitation Research Center, Beijing, China
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China
| | - Tong Zhang
- Department of Neurorehabilitation, China Rehabilitation Research Center, Beijing, China
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China
| | - Lixu Liu
- Department of Neurorehabilitation, China Rehabilitation Research Center, Beijing, China
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China
| | - Lei Shan
- Department of Neurorehabilitation, China Rehabilitation Research Center, Beijing, China
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China
| | - Xueyan Hu
- Department of Neurorehabilitation, China Rehabilitation Research Center, Beijing, China
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China
| | - Lingyu Yang
- Department of Neurorehabilitation, China Rehabilitation Research Center, Beijing, China
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China
| | - Fei Gao
- Department of Neurorehabilitation, China Rehabilitation Research Center, Beijing, China
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China
| | - Wu Xiaoli
- Department of Neurorehabilitation, China Rehabilitation Research Center, Beijing, China
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China
| | - Hanzhi Li
- Department of Neurorehabilitation, China Rehabilitation Research Center, Beijing, China
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China
| |
Collapse
|
18
|
Zhang Z, Wang A, Wang Y, Sun W, Zhou X, Xu Q, Mao L, Zhang J. Canthin-6-Ones: Potential Drugs for Chronic Inflammatory Diseases by Targeting Multiple Inflammatory Mediators. Molecules 2023; 28:3381. [PMID: 37110614 PMCID: PMC10141368 DOI: 10.3390/molecules28083381] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/07/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
Chronic inflammatory disease (CID) is a category of medical conditions that causes recurrent inflammatory attacks in multiple tissues. The occurrence of CID is related to inappropriate immune responses to normal tissue substances and invading microbes due to many factors, such as defects in the immune system and imbalanced regulation of commensal microbes. Thus, effectively keeping the immune-associated cells and their products in check and inhibiting aberrant activation of the immune system is a key strategy for the management of CID. Canthin-6-ones are a subclass of β-carboline alkaloids isolated from a wide range of species. Several emerging studies based on in vitro and in vivo experiments reveal that canthin-6-ones may have potential therapeutic effects on many inflammatory diseases. However, no study has yet summarized the anti-inflammatory functions and the underlying mechanisms of this class of compounds. This review provides an overview of these studies, focusing on the disease entities and the inflammatory mediators that have been shown to be affected by canthin-6-ones. In particular, the major signaling pathways affected by canthin-6-ones, such as the NLR family pyrin domain containing 3 (NLRP3) inflammasome and the NF-κB signaling pathway, and their roles in several CIDs are discussed. Moreover, we discuss the limitations in studies of canthin-6-ones and provide possible solutions. In addition, a perspective that may suggest possible future research directions is provided. This work may be helpful for further mechanistic studies and possible therapeutic applications of canthin-6-ones in the treatment of CID.
Collapse
Affiliation(s)
- Zongying Zhang
- Department of Immunology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, China
| | - Anqi Wang
- Department of Immunology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, China
| | - Yunhan Wang
- Department of Immunology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, China
| | - Weichen Sun
- Department of Immunology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, China
| | - Xiaorong Zhou
- Department of Immunology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, China
| | - Qiuyun Xu
- Department of Immunology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, China
| | - Liming Mao
- Department of Immunology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, China
- Basic Medical Research Center, School of Medicine, Nantong University, Nantong 226019, China
| | - Jie Zhang
- Department of Immunology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, China
| |
Collapse
|
19
|
Trempe JF, Gehring K. Structural mechanisms of mitochondrial quality control mediated by PINK1 and parkin. J Mol Biol 2023:168090. [PMID: 37054910 DOI: 10.1016/j.jmb.2023.168090] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/31/2023] [Accepted: 04/05/2023] [Indexed: 04/15/2023]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease and represents a looming public health crisis as the global population ages. While the etiology of the more common, idiopathic form of the disease remains unknown, the last ten years have seen a breakthrough in our understanding of the genetic forms related to two proteins that regulate a quality control system for the removal of damaged or non-functional mitochondria. Here, we review the structure of these proteins, PINK1, a protein kinase, and parkin, a ubiquitin ligase with an emphasis on the molecular mechanisms responsible for their recognition of dysfunctional mitochondria and control of the subsequent ubiquitination cascade. Recent atomic structures have revealed the basis of PINK1 substrate specificity and the conformational changes responsible for activation of PINK1 and parkin catalytic activity. Progress in understanding the molecular basis of mitochondrial quality control promises to open new avenues for therapeutic interventions in PD.
Collapse
Affiliation(s)
- Jean-François Trempe
- Department of Pharmacology & Therapeutics, McGill University, Montreal, Quebec, Canada; Centre de Recherche en Biologie Structurale
| | - Kalle Gehring
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada; Centre de Recherche en Biologie Structurale
| |
Collapse
|
20
|
Shafique A, Brughera M, Lualdi M, Alberio T. The Role of Rab Proteins in Mitophagy: Insights into Neurodegenerative Diseases. Int J Mol Sci 2023; 24:6268. [PMID: 37047239 PMCID: PMC10094445 DOI: 10.3390/ijms24076268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 03/22/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023] Open
Abstract
Mitochondrial dysfunction and vesicular trafficking alterations have been implicated in the pathogenesis of several neurodegenerative diseases. It has become clear that pathogenetic pathways leading to neurodegeneration are often interconnected. Indeed, growing evidence suggests a concerted contribution of impaired mitophagy and vesicles formation in the dysregulation of neuronal homeostasis, contributing to neuronal cell death. Among the molecular factors involved in the trafficking of vesicles, Ras analog in brain (Rab) proteins seem to play a central role in mitochondrial quality checking and disposal through both canonical PINK1/Parkin-mediated mitophagy and novel alternative pathways. In turn, the lack of proper elimination of dysfunctional mitochondria has emerged as a possible causative/early event in some neurodegenerative diseases. Here, we provide an overview of major findings in recent years highlighting the role of Rab proteins in dysfunctional mitochondrial dynamics and mitophagy, which are characteristic of neurodegenerative diseases. A further effort should be made in the coming years to clarify the sequential order of events and the molecular factors involved in the different processes. A clear cause-effect view of the pathogenetic pathways may help in understanding the molecular basis of neurodegeneration.
Collapse
Affiliation(s)
| | | | | | - Tiziana Alberio
- Department of Science and High Technology, Center of Research in Neuroscience, University of Insubria, I-21052 Busto Arsizio, VA, Italy
| |
Collapse
|
21
|
Heß T, Oehlwein C, Milani TL. Anticipatory Postural Adjustments and Compensatory Postural Responses to Multidirectional Perturbations-Effects of Medication and Subthalamic Nucleus Deep Brain Stimulation in Parkinson's Disease. Brain Sci 2023; 13:brainsci13030454. [PMID: 36979264 PMCID: PMC10046463 DOI: 10.3390/brainsci13030454] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/01/2023] [Accepted: 03/03/2023] [Indexed: 03/30/2023] Open
Abstract
BACKGROUND Postural instability is one of the most restricting motor symptoms for patients with Parkinson's disease (PD). While medication therapy only shows minor effects, it is still unclear whether medication in conjunction with deep brain stimulation (DBS) of the subthalamic nucleus (STN) improves postural stability. Hence, the aim of this study was to investigate whether PD patients treated with medication in conjunction with STN-DBS have superior postural control compared to patients treated with medication alone. METHODS Three study groups were tested: PD patients on medication (PD-MED), PD patients on medication and on STN-DBS (PD-MED-DBS), and healthy elderly subjects (HS) as a reference. Postural performance, including anticipatory postural adjustments (APA) prior to perturbation onset and compensatory postural responses (CPR) following multidirectional horizontal perturbations, was analyzed using force plate and electromyography data. RESULTS Regardless of the treatment condition, both patient groups showed inadequate APA and CPR with early and pronounced antagonistic muscle co-contractions compared to healthy elderly subjects. Comparing the treatment conditions, study group PD-MED-DBS only showed minor advantages over group PD-MED. In particular, group PD-MED-DBS showed faster postural reflexes and tended to have more physiological co-contraction ratios. CONCLUSION medication in conjunction with STN-DBS may have positive effects on the timing and amplitude of postural control.
Collapse
Affiliation(s)
- Tobias Heß
- Department of Human Locomotion, Chemnitz University of Technology, 09126 Chemnitz, Germany
| | - Christian Oehlwein
- Neurological Outpatient Clinic for Parkinson Disease and Deep Brain Stimulation, 07551 Gera, Germany
| | - Thomas L Milani
- Department of Human Locomotion, Chemnitz University of Technology, 09126 Chemnitz, Germany
| |
Collapse
|
22
|
Connelly EM, Frankel KS, Shaw GS. Parkin and mitochondrial signalling. Cell Signal 2023; 106:110631. [PMID: 36803775 DOI: 10.1016/j.cellsig.2023.110631] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023]
Abstract
Aging, toxic chemicals and changes to the cellular environment are sources of oxidative damage to mitochondria which contribute to neurodegenerative conditions including Parkinson's disease. To counteract this, cells have developed signalling mechanisms to identify and remove select proteins and unhealthy mitochondria to maintain homeostasis. Two important proteins that work in concert to control mitochondrial damage are the protein kinase PINK1 and the E3 ligase parkin. In response to oxidative stress, PINK1 phosphorylates ubiquitin present on proteins at the mitochondrial surface. This signals the translocation of parkin, accelerates further phosphorylation, and stimulates ubiquitination of outer mitochondrial membrane proteins such as Miro1/2 and Mfn1/2. The ubiquitination of these proteins is the key step needed to target them for degradation via the 26S proteasomal machinery or eliminate the entire organelle through mitophagy. This review highlights the signalling mechanisms used by PINK1 and parkin and presents several outstanding questions yet to be resolved.
Collapse
Affiliation(s)
- Elizabeth M Connelly
- Department of Biochemistry, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Karling S Frankel
- Department of Biochemistry, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Gary S Shaw
- Department of Biochemistry, The University of Western Ontario, London, Ontario N6A 5C1, Canada.
| |
Collapse
|
23
|
Cell Biology of Parkin: Clues to the Development of New Therapeutics for Parkinson's Disease. CNS Drugs 2022; 36:1249-1267. [PMID: 36378485 DOI: 10.1007/s40263-022-00973-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/20/2022] [Indexed: 11/16/2022]
Abstract
Parkinson's disease is the second most prevalent neurodegenerative disease and contributes significantly to morbidity globally. Currently, no disease-modifying therapies exist to combat this disorder. Insights from the molecular and cellular pathobiology of the disease seems to indicate promising therapeutic targets. The parkin protein has been extensively studied for its role in autosomal recessive Parkinson's disease and, more recently, its role in sporadic Parkinson's disease. Parkin is an E3 ubiquitin ligase that plays a prominent role in mitochondrial quality control, mitochondrial-dependent cell death pathways, and other diverse functions. Understanding the numerous roles of parkin has introduced many new possibilities for therapeutic modalities in treating both autosomal recessive Parkinson's disease and sporadic Parkinson's disease. In this article, we review parkin biology with an emphasis on mitochondrial-related functions and propose novel, potentially disease-modifying therapeutic approaches for treating this debilitating condition.
Collapse
|
24
|
Peng W, Kobeissy F, Mondello S, Barsa C, Mechref Y. MS-based glycomics: An analytical tool to assess nervous system diseases. Front Neurosci 2022; 16:1000179. [PMID: 36408389 PMCID: PMC9671362 DOI: 10.3389/fnins.2022.1000179] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 10/05/2022] [Indexed: 08/27/2023] Open
Abstract
Neurological diseases affect millions of peopleochemistryorldwide and are continuously increasing due to the globe's aging population. Such diseases affect the nervous system and are characterized by a progressive decline in brain function and progressive cognitive impairment, decreasing the quality of life for those with the disease as well as for their families and loved ones. The increased burden of nervous system diseases demands a deeper insight into the biomolecular mechanisms at work during disease development in order to improve clinical diagnosis and drug design. Recently, evidence has related glycosylation to nervous system diseases. Glycosylation is a vital post-translational modification that mediates many biological functions, and aberrant glycosylation has been associated with a variety of diseases. Thus, the investigation of glycosylation in neurological diseases could provide novel biomarkers and information for disease pathology. During the last decades, many techniques have been developed for facilitation of reliable and efficient glycomic analysis. Among these, mass spectrometry (MS) is considered the most powerful tool for glycan analysis due to its high resolution, high sensitivity, and the ability to acquire adequate structural information for glycan identification. Along with MS, a variety of approaches and strategies are employed to enhance the MS-based identification and quantitation of glycans in neurological samples. Here, we review the advanced glycomic tools used in nervous system disease studies, including separation techniques prior to MS, fragmentation techniques in MS, and corresponding strategies. The glycan markers in common clinical nervous system diseases discovered by utilizing such MS-based glycomic tools are also summarized and discussed.
Collapse
Affiliation(s)
- Wenjing Peng
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX, United States
| | - Firas Kobeissy
- Program for Neurotrauma, Neuroproteomics and Biomarkers Research, Department of Emergency Medicine, University of Florida, Gainesville, FL, United States
| | - Stefania Mondello
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Chloe Barsa
- Program for Neurotrauma, Neuroproteomics and Biomarkers Research, Department of Emergency Medicine, University of Florida, Gainesville, FL, United States
| | - Yehia Mechref
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX, United States
| |
Collapse
|
25
|
Sahoo S, Padhy AA, Kumari V, Mishra P. Role of Ubiquitin-Proteasome and Autophagy-Lysosome Pathways in α-Synuclein Aggregate Clearance. Mol Neurobiol 2022; 59:5379-5407. [PMID: 35699874 DOI: 10.1007/s12035-022-02897-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 05/21/2022] [Indexed: 11/26/2022]
Abstract
Synuclein aggregation in neuronal cells is the primary underlying cause of synucleinopathies. Changes in gene expression patterns, structural modifications, and altered interactions with other cellular proteins often trigger aggregation of α-synuclein, which accumulates as oligomers or fibrils in Lewy bodies. Although fibrillar forms of α-synuclein are primarily considered pathological, recent studies have revealed that even the intermediate states of aggregates are neurotoxic, complicating the development of therapeutic interventions. Autophagy and ubiquitin-proteasome pathways play a significant role in maintaining the soluble levels of α-synuclein inside cells; however, the heterogeneous nature of the aggregates presents a significant bottleneck to its degradation by these cellular pathways. With studies focused on identifying the proteins that modulate synuclein aggregation and clearance, detailed mechanistic insights are emerging about the individual and synergistic effects of these degradation pathways in regulating soluble α-synuclein levels. In this article, we discuss the impact of α-synuclein aggregation on autophagy-lysosome and ubiquitin-proteasome pathways and the therapeutic strategies that target various aspects of synuclein aggregation or degradation via these pathways. Additionally, we also highlight the natural and synthetic compounds that have shown promise in alleviating the cellular damage caused due to synuclein aggregation.
Collapse
Affiliation(s)
- Subhashree Sahoo
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, India
| | - Amrita Arpita Padhy
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, India
| | - Varsha Kumari
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, India
| | - Parul Mishra
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, India.
| |
Collapse
|
26
|
Calabrese G, Molzahn C, Mayor T. Protein interaction networks in neurodegenerative diseases: from physiological function to aggregation. J Biol Chem 2022; 298:102062. [PMID: 35623389 PMCID: PMC9234719 DOI: 10.1016/j.jbc.2022.102062] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 04/26/2022] [Accepted: 05/18/2022] [Indexed: 11/25/2022] Open
Abstract
The accumulation of protein inclusions is linked to many neurodegenerative diseases that typically develop in older individuals, due to a combination of genetic and environmental factors. In rare familial neurodegenerative disorders, genes encoding for aggregation-prone proteins are often mutated. While the underlying mechanism leading to these diseases still remains to be fully elucidated, efforts in the past 20 years revealed a vast network of protein–protein interactions that play a major role in regulating the aggregation of key proteins associated with neurodegeneration. Misfolded proteins that can oligomerize and form insoluble aggregates associate with molecular chaperones and other elements of the proteolytic machineries that are the frontline workers attempting to protect the cells by promoting clearance and preventing aggregation. Proteins that are normally bound to aggregation-prone proteins can become sequestered and mislocalized in protein inclusions, leading to their loss of function. In contrast, mutations, posttranslational modifications, or misfolding of aggregation-prone proteins can lead to gain of function by inducing novel or altered protein interactions, which in turn can impact numerous essential cellular processes and organelles, such as vesicle trafficking and the mitochondria. This review examines our current knowledge of protein–protein interactions involving several key aggregation-prone proteins that are associated with Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, or amyotrophic lateral sclerosis. We aim to provide an overview of the protein interaction networks that play a central role in driving or mitigating inclusion formation, while highlighting some of the key proteomic studies that helped to uncover the extent of these networks.
Collapse
Affiliation(s)
- Gaetano Calabrese
- Michael Smith Laboratories, University of British Columbia, V6T 1Z4 Vancouver BC, Canada.
| | - Cristen Molzahn
- Michael Smith Laboratories, University of British Columbia, V6T 1Z4 Vancouver BC, Canada
| | - Thibault Mayor
- Michael Smith Laboratories, University of British Columbia, V6T 1Z4 Vancouver BC, Canada.
| |
Collapse
|
27
|
An Amish founder population reveals rare-population genetic determinants of the human lipidome. Commun Biol 2022; 5:334. [PMID: 35393526 PMCID: PMC8989972 DOI: 10.1038/s42003-022-03291-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 03/17/2022] [Indexed: 12/02/2022] Open
Abstract
Identifying the genetic determinants of inter-individual variation in lipid species (lipidome) may provide deeper understanding and additional insight into the mechanistic effect of complex lipidomic pathways in CVD risk and progression beyond simple traditional lipids. Previous studies have been largely population based and thus only powered to discover associations with common genetic variants. Founder populations represent a powerful resource to accelerate discovery of previously unknown biology associated with rare population alleles that have risen to higher frequency due to genetic drift. We performed a genome-wide association scan of 355 lipid species in 650 individuals from the Amish founder population including 127 lipid species not previously tested. To the best of our knowledge, we report for the first time the lipid species associated with two rare-population but Amish-enriched lipid variants: APOB_rs5742904 and APOC3_rs76353203. We also identified novel associations for 3 rare-population Amish-enriched loci with several sphingolipids and with proposed potential functional/causal variant in each locus including GLTPD2_rs536055318, CERS5_rs771033566, and AKNA_rs531892793. We replicated 7 previously known common loci including novel associations with two sterols: androstenediol with UGT locus and estriol with SLC22A8/A24 locus. Our results show the double power of founder populations and detailed lipidome to discover novel trait-associated variants. A GWAS of 355 lipid species in the Old Order Amish founder population reveals associations between Amish-enriched loci and several sphingolipids.
Collapse
|
28
|
The Role of NEDD4 E3 Ubiquitin–Protein Ligases in Parkinson’s Disease. Genes (Basel) 2022; 13:genes13030513. [PMID: 35328067 PMCID: PMC8950476 DOI: 10.3390/genes13030513] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 03/03/2022] [Indexed: 01/25/2023] Open
Abstract
Parkinson’s disease (PD) is a debilitating neurodegenerative disease that causes a great clinical burden. However, its exact molecular pathologies are not fully understood. Whilst there are a number of avenues for research into slowing, halting, or reversing PD, one central idea is to enhance the clearance of the proposed aetiological protein, oligomeric α-synuclein. Oligomeric α-synuclein is the main constituent protein in Lewy bodies and neurites and is considered neurotoxic. Multiple E3 ubiquitin-protein ligases, including the NEDD4 (neural precursor cell expressed developmentally downregulated protein 4) family, parkin, SIAH (mammalian homologues of Drosophila seven in absentia), CHIP (carboxy-terminus of Hsc70 interacting protein), and SCFFXBL5 SCF ubiquitin ligase assembled by the S-phase kinase-associated protein (SKP1), cullin-1 (Cul1), a zinc-binding RING finger protein, and the F-box domain/Leucine-rich repeat protein 5-containing protein FBXL5), have been shown to be able to ubiquitinate α-synuclein, influencing its subsequent degradation via the proteasome or lysosome. Here, we explore the link between NEDD4 ligases and PD, which is not only via α-synuclein but further strengthened by several additional substrates and interaction partners. Some members of the NEDD4 family of ligases are thought to crosstalk even with PD-related genes and proteins found to be mutated in familial forms of PD. Mutations in NEDD4 family genes have not been observed in PD patients, most likely because of their essential survival function during development. Following further in vivo studies, it has been thought that NEDD4 ligases may be viable therapeutic targets in PD. NEDD4 family members could clear toxic proteins, enhancing cell survival and slowing disease progression, or might diminish beneficial proteins, reducing cell survival and accelerating disease progression. Here, we review studies to date on the expression and function of NEDD4 ubiquitin ligases in the brain and their possible impact on PD pathology.
Collapse
|
29
|
Caplliure-Llopis J, Escrivá D, Navarro-Illana E, Benlloch M, de la Rubia Ortí JE, Barrios C. Bone Quality in Patients with Parkinson's Disease Determined by Quantitative Ultrasound (QUS) of the Calcaneus: Influence of Sex Differences. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:2804. [PMID: 35270499 PMCID: PMC8910506 DOI: 10.3390/ijerph19052804] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 02/04/2023]
Abstract
(1) Background: Parkinson’s disease (PD) is a relatively common neurodegenerative disease in elderly individuals, with a high risk of falls. There is abundant literature on the relationship between PD and osteoporosis. The aim of this study is to describe the bone quality of a population with PD by calcaneal ultrasound and to compare it with a healthy control, assessing the influence of possible sex differences. (2) Methods: 21 patients diagnosed with PD were recruited. The control group was composed of 30 healthy individuals with similar sociodemographic characteristics. The bone quality of all participants was assessed using calcaneal quantitative ultrasound (QUS). The parameters recorded were broadband ultrasound attenuation (BUA, in decibels per megahertz), imaging speed of sound (SOS, in meters per second), stiffness index (SI) and T-score of each participant. Bone mineral density (BMD) was estimated using the equation BMD = 0.002592 × (BUA + SOS) − 3.687 (g/cm2). (3) Results: significant differences were observed between the healthy control and the PD group: the T-score was lower in the PD group (p < 0.05) and SOS was higher in Parkinson’s disease patients (p < 0.05), while 28.6% of the PD patients were osteoporotic with T-score values lower than −1.5 compared to 16.7% of osteoporotic individuals in the control group (p < 0.01). Regarding the sex, there were significant differences (p < 0.05) between the females of the PD group vs. control group, showing a significant difference in the SI (71.4 ± 14.7 vs. 87.8 ± 12), T-score (−2.19 ± 1.1 vs. −0.15 ± 0.8), BUA (104.5 ± 13 vs. 116 ± 10.6) and BMD (0.49 ± 0.09 vs. 0.60 ± 0.08), with no difference in the comparison between the male groups; and the comparison between both sexes in T-score only showed significant differences for the PD group (p < 0.05), with worse bone quality in women. (4) Conclusions: this study shows poorer bone quality in female patients with PD, who have a higher percentage of osteoporosis than healthy patients. The QUS technique of the calcaneus seems adequate for these determinations in patients with Parkinson’s disease.
Collapse
Affiliation(s)
- Jordi Caplliure-Llopis
- Institute for Research on Musculoskeletal Disorders, Valencia Catholic University San Vincente Martir, Quevedo 2, 46001 Valencia, Spain; (J.C.-L.); (D.E.); (C.B.)
- Department of Primary Care, Hospital Universitario de La Ribera, 46600 Valencia, Spain
| | - Dolores Escrivá
- Institute for Research on Musculoskeletal Disorders, Valencia Catholic University San Vincente Martir, Quevedo 2, 46001 Valencia, Spain; (J.C.-L.); (D.E.); (C.B.)
- Department of Basic Medical Sciences, Catholic University of Valencia San Vicente Martir, 46001 Valencia, Spain;
| | - Esther Navarro-Illana
- Department of Basic Medical Sciences, Catholic University of Valencia San Vicente Martir, 46001 Valencia, Spain;
| | - María Benlloch
- Department of Basic Medical Sciences, Catholic University of Valencia San Vicente Martir, 46001 Valencia, Spain;
| | | | - Carlos Barrios
- Institute for Research on Musculoskeletal Disorders, Valencia Catholic University San Vincente Martir, Quevedo 2, 46001 Valencia, Spain; (J.C.-L.); (D.E.); (C.B.)
| |
Collapse
|
30
|
Kwon DH, Hwang JS, Kim SG, Jang YE, Shin TH, Lee G. Cerebrospinal Fluid Metabolome in Parkinson's Disease and Multiple System Atrophy. Int J Mol Sci 2022; 23:ijms23031879. [PMID: 35163800 PMCID: PMC8836409 DOI: 10.3390/ijms23031879] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/04/2022] [Accepted: 02/04/2022] [Indexed: 11/16/2022] Open
Abstract
Parkinson’s disease (PD) and multiple system atrophy (MSA) belong to the neurodegenerative group of synucleinopathies; differential diagnosis between PD and MSA is difficult, especially at early stages, owing to their clinical and biological similarities. Thus, there is a pressing need to identify metabolic biomarkers for these diseases. The metabolic profile of the cerebrospinal fluid (CSF) is reported to be altered in PD and MSA; however, the altered metabolites remain unclear. We created a single network with altered metabolites in PD and MSA based on the literature and assessed biological functions, including metabolic disorders of the nervous system, inflammation, concentration of ATP, and neurological disorder, through bioinformatics methods. Our in-silico prediction-based metabolic networks are consistent with Parkinsonism events. Although metabolomics approaches provide a more quantitative understanding of biochemical events underlying the symptoms of PD and MSA, limitations persist in covering molecules related to neurodegenerative disease pathways. Thus, omics data, such as proteomics and microRNA, help understand the altered metabolomes mechanism. In particular, integrated omics and machine learning approaches will be helpful to elucidate the pathological mechanisms of PD and MSA. This review discusses the altered metabolites between PD and MSA in the CSF and omics approaches to discover diagnostic biomarkers.
Collapse
Affiliation(s)
- Do Hyeon Kwon
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea; (D.H.K.); (J.S.H.); (S.G.K.); (Y.E.J.)
| | - Ji Su Hwang
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea; (D.H.K.); (J.S.H.); (S.G.K.); (Y.E.J.)
| | - Seok Gi Kim
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea; (D.H.K.); (J.S.H.); (S.G.K.); (Y.E.J.)
| | - Yong Eun Jang
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea; (D.H.K.); (J.S.H.); (S.G.K.); (Y.E.J.)
| | - Tae Hwan Shin
- Department of Physiology, Ajou University School of Medicine, Suwon 16499, Korea
- Correspondence: (T.H.S.); (G.L.)
| | - Gwang Lee
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea; (D.H.K.); (J.S.H.); (S.G.K.); (Y.E.J.)
- Department of Physiology, Ajou University School of Medicine, Suwon 16499, Korea
- Correspondence: (T.H.S.); (G.L.)
| |
Collapse
|
31
|
Nag S, Jahan M, Tóth M, Nakao R, Varrone A, Halldin C. PET Imaging of VMAT2 with the Novel Radioligand [ 18F]FE-DTBZ-d4 in Nonhuman Primates: Comparison with [ 11C]DTBZ and [ 18F]FE-DTBZ. ACS Chem Neurosci 2021; 12:4580-4586. [PMID: 34813272 PMCID: PMC8678981 DOI: 10.1021/acschemneuro.1c00651] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
![]()
The vesicular monoamine
transporter type 2 (VMAT2) is believed
to be responsible for the uptake of monoamines into the vesicles of
the synaptic terminals. Two VMAT2 radioligands [11C]DTBZ
and [18F]FP-DTBZ have been used to assess the degree of
nigrostriatal deficit in Parkinson’s disease (PD) using positron
emission tomography (PET). [18F]FE-DTBZ-d4, the nondeuterated
analogue of [18F]FE-DTBZ showed similar imaging properties
with better stability against defluorination. Therefore, [18F]FE-DTBZ-d4 draws attention to be investigated as an imaging marker
for VMAT2 in the brain. The aim of this study was to investigate the
brain kinetics and quantification of [18F]FE-DTBZ-d4 in
nonhuman primates (NHPs), with comparison to [11C]DTBZ
and [18F]FE-DTBZ. Radiolabeling was successfully achieved
either by one-step 11C-methylation or by a two-step fluorine-18
nucleophilic substitution reaction. The stability and radiochemical
yield were analyzed with high-performance liquid chromatography (HPLC).
Three female cynomolgus monkeys were included in the study and underwent
a total of 12 positron emission tomography (PET) measurements. Each
monkey was examined with each tracer. In addition, two pretreatment
and one displacement PET measurements with tetrabenazine (2.0 mg/kg)
were performed for [18F]FE-DTBZ-d4. All PET measurements
were conducted using a high-resolution research tomograph (HRRT) system.
Radiometabolites were measured in monkey plasma using gradient radio-HPLC.
[18F]FE-DTBZ-d4 (SUV: 4.28 ± 1.01) displayed higher
brain uptake compared to both [18F]FE-DTBZ (SUV: 3.43 ±
0.54) and [11C]DTBZ (SUV: 3.06 ± 0.32) and faster
washout. Binding potential (BPND) values of [18F]FE-DTBZ-d4 in different brain regions (putamen: 5.5 ± 1.4;
caudate: 4.4 ± 1.1; midbrain: 1.4 ± 0.4) were higher than
those of [11C]DTBZ and [18F]FE-DTBZ. [18F]FE-DTBZ showed faster radiometabolism in plasma compared to [11C]DTBZ and [18F]FE-DTBZ-d4. [18F]FE-DTBZ-d4
is a suitable radioligand for quantification of VMAT2 in the nonhuman
primate brain, with better imaging properties than [11C]DTBZ
and [18F]FE-DTBZ. A preliminary comparison suggests that
[18F]FE-DTBZ-d4 has increased stability against defluorination
compared to the nondeuterated analogue.
Collapse
Affiliation(s)
- Sangram Nag
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm 17176, Sweden
| | - Mahabuba Jahan
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm 17176, Sweden
| | - Miklós Tóth
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm 17176, Sweden
| | - Ryuji Nakao
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm 17176, Sweden
| | - Andrea Varrone
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm 17176, Sweden
| | - Christer Halldin
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm 17176, Sweden
| |
Collapse
|
32
|
The role of amyloids in Alzheimer's and Parkinson's diseases. Int J Biol Macromol 2021; 190:44-55. [PMID: 34480905 DOI: 10.1016/j.ijbiomac.2021.08.197] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 08/23/2021] [Accepted: 08/26/2021] [Indexed: 11/23/2022]
Abstract
With varying clinical symptoms, most neurodegenerative diseases are associated with abnormal loss of neurons. They share the same common pathogenic mechanisms involving misfolding and aggregation, and these visible aggregates of proteins are deposited in the central nervous system. Amyloid formation is thought to arise from partial unfolding of misfolded proteins leading to the exposure of hydrophobic surfaces, which interact with other similar structures and give rise to form dimers, oligomers, protofibrils, and eventually mature fibril aggregates. Accumulating evidence indicates that amyloid oligomers, not amyloid fibrils, are the most toxic species that causes Alzheimer's disease (AD) and Parkinson's disease (PD). AD has recently been recognized as the 'twenty-first century plague', with an incident rate of 1% at 60 years of age, which then doubles every fifth year. Currently, 5.3 million people in the US are afflicted with this disease, and the number of cases is expected to rise to 13.5 million by 2050. PD, a disorder of the brain, is the second most common form of dementia, characterized by difficulty in walking and movement. Keeping the above views in mind, in this review we have focused on the roles of amyloid in neurodegenerative diseases including AD and PD, the involvement of amyloid in mitochondrial dysfunction leading to neurodegeneration, are also considered in the review.
Collapse
|
33
|
Pirooznia SK, Rosenthal LS, Dawson VL, Dawson TM. Parkinson Disease: Translating Insights from Molecular Mechanisms to Neuroprotection. Pharmacol Rev 2021; 73:33-97. [PMID: 34663684 DOI: 10.1124/pharmrev.120.000189] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Parkinson disease (PD) used to be considered a nongenetic condition. However, the identification of several autosomal dominant and recessive mutations linked to monogenic PD has changed this view. Clinically manifest PD is then thought to occur through a complex interplay between genetic mutations, many of which have incomplete penetrance, and environmental factors, both neuroprotective and increasing susceptibility, which variably interact to reach a threshold over which PD becomes clinically manifested. Functional studies of PD gene products have identified many cellular and molecular pathways, providing crucial insights into the nature and causes of PD. PD originates from multiple causes and a range of pathogenic processes at play, ultimately culminating in nigral dopaminergic loss and motor dysfunction. An in-depth understanding of these complex and possibly convergent pathways will pave the way for therapeutic approaches to alleviate the disease symptoms and neuroprotective strategies to prevent disease manifestations. This review is aimed at providing a comprehensive understanding of advances made in PD research based on leveraging genetic insights into the pathogenesis of PD. It further discusses novel perspectives to facilitate identification of critical molecular pathways that are central to neurodegeneration that hold the potential to develop neuroprotective and/or neurorestorative therapeutic strategies for PD. SIGNIFICANCE STATEMENT: A comprehensive review of PD pathophysiology is provided on the complex interplay of genetic and environmental factors and biologic processes that contribute to PD pathogenesis. This knowledge identifies new targets that could be leveraged into disease-modifying therapies to prevent or slow neurodegeneration in PD.
Collapse
Affiliation(s)
- Sheila K Pirooznia
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering (S.K.P., V.L.D., T.M.D.), Departments of Neurology (S.K.P., L.S.R., V.L.D., T.M.D.), Departments of Physiology (V.L.D.), Solomon H. Snyder Department of Neuroscience (V.L.D., T.M.D.), Department of Pharmacology and Molecular Sciences (T.M.D.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.); and Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.)
| | - Liana S Rosenthal
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering (S.K.P., V.L.D., T.M.D.), Departments of Neurology (S.K.P., L.S.R., V.L.D., T.M.D.), Departments of Physiology (V.L.D.), Solomon H. Snyder Department of Neuroscience (V.L.D., T.M.D.), Department of Pharmacology and Molecular Sciences (T.M.D.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.); and Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.)
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering (S.K.P., V.L.D., T.M.D.), Departments of Neurology (S.K.P., L.S.R., V.L.D., T.M.D.), Departments of Physiology (V.L.D.), Solomon H. Snyder Department of Neuroscience (V.L.D., T.M.D.), Department of Pharmacology and Molecular Sciences (T.M.D.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.); and Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.)
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering (S.K.P., V.L.D., T.M.D.), Departments of Neurology (S.K.P., L.S.R., V.L.D., T.M.D.), Departments of Physiology (V.L.D.), Solomon H. Snyder Department of Neuroscience (V.L.D., T.M.D.), Department of Pharmacology and Molecular Sciences (T.M.D.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.); and Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.)
| |
Collapse
|
34
|
Caminiti SP, Carli G, Avenali M, Blandini F, Perani D. Clinical and Dopamine Transporter Imaging Trajectories in a Cohort of Parkinson's Disease Patients with GBA Mutations. Mov Disord 2021; 37:106-118. [PMID: 34596920 DOI: 10.1002/mds.28818] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 09/08/2021] [Accepted: 09/14/2021] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Glucosylceramidase (GBA) mutations are considered the most common genetic risk factors for developing Parkinson's disease (PD). OBJECTIVES We aimed to assess, at different time points, the integrity of brain striatal and extra-striatal dopamine pathways and clinical phenotype of a group of PD subjects bearing heterozygous GBA mutations (GBA-PD), compared with a group of idiopathic PD patients (iPD) stratified by age at disease onset. A longitudinal approach was adopted to evaluate the progression over time for clinical and 123 I-FP-CIT SPECT imaging features. METHODS We considered 46 GBA-PD patients and 339 iPD patients, subdivided into two groups according to age at PD onset (n = 58 < 50 years and n = 281 > 50 years). We measured differences in the occurrence/severity/progression of motor and non-motor features, 123 I-FP-CIT standard uptake value ratios (SUVr) in striatal and extra-striatal regions, and global cognitive deterioration over time in a subset of 168 cases with available follow-up. RESULTS At baseline, the GBA-PD cohort showed more severe motor and cognitive deficits than the early-iPD cohort. The 123 I-FP-CIT SUVr reduction in the striatal and the extra-striatal regions was more marked in the GBA-PD than the early- and late-iPD cohorts. Both GBA-PD and late-iPD patients had a significant annual deterioration in their global cognitive performance, while the early-iPD group showed global cognitive stability over time. At follow-up, the iPD cohorts became similar to the GBA-PD group in 123 I-FP-CIT SUVr reduction. CONCLUSION These new findings support the hypothesis of a biological role of GBA mutations in accelerating the early neurodegenerative processes in PD, leading to the malignant clinical phenotype. © 2021 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Silvia Paola Caminiti
- School of Psychology, Vita-Salute San Raffaele University, Milan, Italy.,In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Nuclear Medicine Unit, San Raffaele Hospital, Milan, Italy
| | - Giulia Carli
- School of Psychology, Vita-Salute San Raffaele University, Milan, Italy.,In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Micol Avenali
- IRCCS Mondino Foundation, Pavia, Italy.,Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy
| | - Fabio Blandini
- IRCCS Mondino Foundation, Pavia, Italy.,Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy
| | - Daniela Perani
- School of Psychology, Vita-Salute San Raffaele University, Milan, Italy.,In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Nuclear Medicine Unit, San Raffaele Hospital, Milan, Italy
| |
Collapse
|
35
|
Curcuma longa extract ameliorates motor and cognitive deficits of 6-hydroxydopamine-infused Parkinson’s disease model rats. ADVANCES IN TRADITIONAL MEDICINE 2021. [DOI: 10.1007/s13596-021-00606-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
36
|
Fonseca Cabral G, Schaan AP, Cavalcante GC, Sena-dos-Santos C, de Souza TP, Souza Port’s NM, dos Santos Pinheiro JA, Ribeiro-dos-Santos Â, Vidal AF. Nuclear and Mitochondrial Genome, Epigenome and Gut Microbiome: Emerging Molecular Biomarkers for Parkinson's Disease. Int J Mol Sci 2021; 22:9839. [PMID: 34576000 PMCID: PMC8471599 DOI: 10.3390/ijms22189839] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 06/23/2021] [Accepted: 06/28/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Parkinson's disease (PD) is currently the second most common neurodegenerative disorder, burdening about 10 million elderly individuals worldwide. The multifactorial nature of PD poses a difficult obstacle for understanding the mechanisms involved in its onset and progression. Currently, diagnosis depends on the appearance of clinical signs, some of which are shared among various neurologic disorders, hindering early diagnosis. There are no effective tools to prevent PD onset, detect the disease in early stages or accurately report the risk of disease progression. Hence, there is an increasing demand for biomarkers that may identify disease onset and progression, as treatment-based medicine may not be the best approach for PD. Over the last few decades, the search for molecular markers to predict susceptibility, aid in accurate diagnosis and evaluate the progress of PD have intensified, but strategies aimed to improve individualized patient care have not yet been established. CONCLUSIONS Genomic variation, regulation by epigenomic mechanisms, as well as the influence of the host gut microbiome seem to have a crucial role in the onset and progress of PD, thus are considered potential biomarkers. As such, the human nuclear and mitochondrial genome, epigenome, and the host gut microbiome might be the key elements to the rise of personalized medicine for PD patients.
Collapse
Affiliation(s)
- Gleyce Fonseca Cabral
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, R. Augusto Correa, Belém 66075-110, Brazil; (G.F.C.); (A.P.S.); (G.C.C.); (C.S.-d.-S.); (T.P.d.S.); (J.A.d.S.P.)
| | - Ana Paula Schaan
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, R. Augusto Correa, Belém 66075-110, Brazil; (G.F.C.); (A.P.S.); (G.C.C.); (C.S.-d.-S.); (T.P.d.S.); (J.A.d.S.P.)
| | - Giovanna C. Cavalcante
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, R. Augusto Correa, Belém 66075-110, Brazil; (G.F.C.); (A.P.S.); (G.C.C.); (C.S.-d.-S.); (T.P.d.S.); (J.A.d.S.P.)
| | - Camille Sena-dos-Santos
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, R. Augusto Correa, Belém 66075-110, Brazil; (G.F.C.); (A.P.S.); (G.C.C.); (C.S.-d.-S.); (T.P.d.S.); (J.A.d.S.P.)
| | - Tatiane Piedade de Souza
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, R. Augusto Correa, Belém 66075-110, Brazil; (G.F.C.); (A.P.S.); (G.C.C.); (C.S.-d.-S.); (T.P.d.S.); (J.A.d.S.P.)
| | - Natacha M. Souza Port’s
- Laboratório de Neurofarmacologia Molecular, Universidade de São Paulo, São Paulo 05508-000, Brazil;
| | - Jhully Azevedo dos Santos Pinheiro
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, R. Augusto Correa, Belém 66075-110, Brazil; (G.F.C.); (A.P.S.); (G.C.C.); (C.S.-d.-S.); (T.P.d.S.); (J.A.d.S.P.)
| | - Ândrea Ribeiro-dos-Santos
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, R. Augusto Correa, Belém 66075-110, Brazil; (G.F.C.); (A.P.S.); (G.C.C.); (C.S.-d.-S.); (T.P.d.S.); (J.A.d.S.P.)
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará–R. dos Mundurucus, Belém 66073-000, Brazil
- Programa de Pós-Graduação em Genética e Biologia Molecular, Universidade Federal do Pará, R. Augusto Correa, Belém 66075-110, Brazil
| | - Amanda F. Vidal
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, R. Augusto Correa, Belém 66075-110, Brazil; (G.F.C.); (A.P.S.); (G.C.C.); (C.S.-d.-S.); (T.P.d.S.); (J.A.d.S.P.)
- Programa de Pós-Graduação em Genética e Biologia Molecular, Universidade Federal do Pará, R. Augusto Correa, Belém 66075-110, Brazil
- ITVDS—Instituto Tecnológico Vale Desenvolvimento Sustentável–R. Boaventura da Silva, Belém 66055-090, Brazil
| |
Collapse
|
37
|
Do HA, Baek KH. Cellular functions regulated by deubiquitinating enzymes in neurodegenerative diseases. Ageing Res Rev 2021; 69:101367. [PMID: 34023421 DOI: 10.1016/j.arr.2021.101367] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 05/07/2021] [Accepted: 05/17/2021] [Indexed: 12/24/2022]
Abstract
Neurodegenerative diseases are one of the most common diseases in mankind. Although there are reports of several candidates that cause neurodegenerative diseases, the exact mechanism of pathogenesis is poorly understood. The ubiquitin-proteasome system (UPS) is an important posttranslational modification for protein degradation and control of homeostasis. Enzymes such as E1, E2, E3 ligases, and deubiquitinating enzymes (DUBs) participating in UPS, regulate disease-inducing proteins by controlling the degree of ubiquitination. Therefore, the development of treatments targeting enzymes for degenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and amyotrophic lateral sclerosis (ALS), is emerging as an attractive perspective. In particular, as DUBs are able to regulate one or more degenerative disease-related proteins, the potential as a therapeutic target is even more evident. DUBs influence the regulation of toxic proteins that cause neurodegenerative diseases by not only their removal, but also by regulating signals associated with mitophagy, autophagy, and endoplasmic reticulum-associated degradation (ERAD). In this review, we analyze not only the cellular processes of DUBs, which control neurodegenerative disease-inducing proteins, but also their potentials as a therapeutic agent for neurodegenerative diseases.
Collapse
|
38
|
Jo J, Yang L, Tran HD, Yu W, Sun AX, Chang YY, Jung BC, Lee SJ, Saw TY, Xiao B, Khoo ATT, Yaw LP, Xie JJ, Lokman H, Ong WY, Lim GGY, Lim KL, Tan EK, Ng HH, Je HS. Lewy Body-like Inclusions in Human Midbrain Organoids Carrying Glucocerebrosidase and α-Synuclein Mutations. Ann Neurol 2021; 90:490-505. [PMID: 34288055 PMCID: PMC9543721 DOI: 10.1002/ana.26166] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 07/12/2021] [Accepted: 07/12/2021] [Indexed: 01/02/2023]
Abstract
Objective We utilized human midbrain‐like organoids (hMLOs) generated from human pluripotent stem cells carrying glucocerebrosidase gene (GBA1) and α‐synuclein (α‐syn; SNCA) perturbations to investigate genotype‐to‐phenotype relationships in Parkinson disease, with the particular aim of recapitulating α‐syn– and Lewy body–related pathologies and the process of neurodegeneration in the hMLO model. Methods We generated and characterized hMLOs from GBA1−/− and SNCA overexpressing isogenic embryonic stem cells and also generated Lewy body–like inclusions in GBA1/SNCA dual perturbation hMLOs and conduritol‐b‐epoxide–treated SNCA triplication hMLOs. Results We identified for the first time that the loss of glucocerebrosidase, coupled with wild‐type α‐syn overexpression, results in a substantial accumulation of detergent‐resistant, β‐sheet–rich α‐syn aggregates and Lewy body–like inclusions in hMLOs. These Lewy body–like inclusions exhibit a spherically symmetric morphology with an eosinophilic core, containing α‐syn with ubiquitin, and can also be formed in Parkinson disease patient–derived hMLOs. We also demonstrate that impaired glucocerebrosidase function promotes the formation of Lewy body–like inclusions in hMLOs derived from patients carrying the SNCA triplication. Interpretation Taken together, the data indicate that our hMLOs harboring 2 major risk factors (glucocerebrosidase deficiency and wild‐type α‐syn overproduction) of Parkinson disease provide a tractable model to further elucidate the underlying mechanisms for progressive Lewy body formation. ANN NEUROL 2021;90:490–505
Collapse
Affiliation(s)
- Junghyun Jo
- Genome Institute of Singapore, Singapore, Singapore.,Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Lin Yang
- Genome Institute of Singapore, Singapore, Singapore
| | - Hoang-Dai Tran
- Genome Institute of Singapore, Singapore, Singapore.,National Neuroscience Institute, Singapore, Singapore
| | - Weonjin Yu
- Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore, Singapore.,Department of Physiology, Seoul National University College of Medicine, Seoul, South Korea
| | - Alfred Xuyang Sun
- Genome Institute of Singapore, Singapore, Singapore.,National Neuroscience Institute, Singapore, Singapore.,Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Ya Yin Chang
- National Neuroscience Institute, Singapore, Singapore
| | - Byung Chul Jung
- Department of Biomedical Sciences, Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, South Korea.,Department of Biomedical Laboratory Science, Masan University, Changwon-si, South Korea
| | - Seung-Jae Lee
- Department of Biomedical Sciences, Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | | | - Bin Xiao
- National Neuroscience Institute, Singapore, Singapore
| | - Audrey Tze Ting Khoo
- Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Lai-Ping Yaw
- Genome Institute of Singapore, Singapore, Singapore
| | | | - Hidayat Lokman
- Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Wei-Yi Ong
- Department of Anatomy, National University of Singapore, Singapore, Singapore
| | | | - Kah-Leong Lim
- National Neuroscience Institute, Singapore, Singapore.,Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Eng-King Tan
- National Neuroscience Institute, Singapore, Singapore
| | - Huck-Hui Ng
- Genome Institute of Singapore, Singapore, Singapore.,Department of Biochemistry, National University of Singapore, Singapore, Singapore.,Department of Biological Sciences, National University of Singapore, Singapore, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Hyunsoo Shawn Je
- Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
39
|
Historical perspective and progress on protein ubiquitination at glutamatergic synapses. Neuropharmacology 2021; 196:108690. [PMID: 34197891 DOI: 10.1016/j.neuropharm.2021.108690] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 06/07/2021] [Accepted: 06/22/2021] [Indexed: 12/23/2022]
Abstract
Transcription-translation coupling leads to the production of proteins that are key for controlling essential neuronal processes that include neuronal development and changes in synaptic strength. Although these events have been a prevailing theme in neuroscience, the regulation of proteins via posttranslational signaling pathways are equally relevant for these neuronal processes. Ubiquitin is one type of posttranslational modification that covalently attaches to its targets/substrates. Ubiquitination of proteins play a key role in multiple signaling pathways, the predominant being removal of its substrates by a large molecular machine called the proteasome. Here, I review 40 years of progress on ubiquitination in the nervous system at glutamatergic synapses focusing on axon pathfinding, synapse formation, presynaptic release, dendritic spine formation, and regulation of postsynaptic glutamate receptors. Finally, I elucidate emerging themes in ubiquitin biology that may challenge our current understanding of ubiquitin signaling in the nervous system.
Collapse
|
40
|
α-synuclein pathogenesis in hiPSC models of Parkinson's disease. Neuronal Signal 2021; 5:NS20210021. [PMID: 34239711 PMCID: PMC8222967 DOI: 10.1042/ns20210021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 06/08/2021] [Accepted: 06/14/2021] [Indexed: 12/17/2022] Open
Abstract
α-synuclein is an increasingly prominent player in the pathology of a variety of neurodegenerative conditions. Parkinson’s disease (PD) is a neurodegenerative disorder that affects mainly the dopaminergic (DA) neurons in the substantia nigra of the brain. Typical of PD pathology is the finding of protein aggregations termed ‘Lewy bodies’ in the brain regions affected. α-synuclein is implicated in many disease states including dementia with Lewy bodies (DLB) and Alzheimer’s disease. However, PD is the most common synucleinopathy and continues to be a significant focus of PD research in terms of the α-synuclein Lewy body pathology. Mutations in several genes are associated with PD development including SNCA, which encodes α-synuclein. A variety of model systems have been employed to study α-synuclein physiology and pathophysiology in an attempt to relate more closely to PD pathology. These models include cellular and animal system exploring transgenic technologies, viral vector expression and knockdown approaches, and models to study the potential prion protein-like effects of α-synuclein. The current review focuses on human induced pluripotent stem cell (iPSC) models with a specific focus on mutations or multiplications of the SNCA gene. iPSCs are a rapidly evolving technology with huge promise in the study of normal physiology and disease modeling in vitro. The ability to maintain a patient’s genetic background and replicate similar cell phenotypes make iPSCs a powerful tool in the study of neurological diseases. This review focuses on the current knowledge about α-synuclein physiological function as well as its role in PD pathogenesis based on human iPSC models.
Collapse
|
41
|
Murillo-González FE, García-Aguilar R, Vega L, Elizondo G. Regulation of Parkin expression as the key balance between neural survival and cancer cell death. Biochem Pharmacol 2021; 190:114650. [PMID: 34111426 DOI: 10.1016/j.bcp.2021.114650] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/02/2021] [Accepted: 06/04/2021] [Indexed: 11/25/2022]
Abstract
Parkin is a cytosolic E3 ubiquitin ligase that plays an important role in neuroprotection by targeting several proteins to be degraded by the 26S proteasome. Its dysfunction has been associated not only with Parkinson's disease (PD) but also with other neurodegenerative pathologies, such as Alzheimer's disease and Huntington's disease. More recently, Parkin has been identified as a tumor suppressor gene implicated in cancer development. Due to the important roles that this E3 ubiquitin ligase plays in cellular homeostasis, its expression, activity, and turnover are tightly regulated. Several reviews have addressed Parkin regulation; however, genetic and epigenetic regulation have been excluded. In addition to posttranslational modifications (PTMs), this review examines the regulatory mechanisms that control Parkin function through gene expression, epigenetic regulation, and degradation. Furthermore, the consequences of disrupting these regulatory processes on human health are discussed.
Collapse
Affiliation(s)
| | | | - Libia Vega
- Department of Toxicology, CINVESTAV-IPN, Av. IPN 2508, C.P. 07360 Mexico City, Mexico
| | - Guillermo Elizondo
- Department of Cellular Biology, CINVESTAV-IPN, Av. IPN 2508, C.P. 07360 Mexico City, Mexico.
| |
Collapse
|
42
|
Carvajal-Oliveros A, Domínguez-Baleón C, Zárate RV, Campusano JM, Narváez-Padilla V, Reynaud E. Nicotine suppresses Parkinson's disease like phenotypes induced by Synphilin-1 overexpression in Drosophila melanogaster by increasing tyrosine hydroxylase and dopamine levels. Sci Rep 2021; 11:9579. [PMID: 33953275 PMCID: PMC8099903 DOI: 10.1038/s41598-021-88910-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 03/30/2021] [Indexed: 11/12/2022] Open
Abstract
It has been observed that there is a lower Parkinson's disease (PD) incidence in tobacco users. Nicotine is a cholinergic agonist and is the principal psychoactive compound in tobacco linked to cigarette addiction. Different studies have shown that nicotine has beneficial effects on sporadic and genetic models of PD. In this work we evaluate nicotine's protective effect in a Drosophila melanogaster model for PD where Synphilin-1 (Sph-1) is expressed in dopaminergic neurons. Nicotine has a moderate effect on dopaminergic neuron survival that becomes more evident as flies age. Nicotine is beneficial on fly survival and motility increasing tyrosine hydroxylase and dopamine levels, suggesting that cholinergic agonists may promote survival and metabolic function of the dopaminergic neurons that express Sph-1. The Sph-1 expressing fly is a good model for the study of early-onset phenotypes such as olfaction loss one of the main non-motor symptom related to PD. Our data suggest that nicotine is an interesting therapeutic molecule whose properties should be explored in future research on the phenotypic modulators of the disease and for the development of new treatments.
Collapse
Affiliation(s)
- Angel Carvajal-Oliveros
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, UNAM, A.P. 510-3, 62210, Cuernavaca, Mor., Mexico
| | - Carmen Domínguez-Baleón
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, UNAM, A.P. 510-3, 62210, Cuernavaca, Mor., Mexico
| | - Rafaella V Zárate
- Laboratorio Neurogenética de la Conducta, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jorge M Campusano
- Laboratorio Neurogenética de la Conducta, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Verónica Narváez-Padilla
- Centro de Investigación en Dinámica Celular, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, Mexico
| | - Enrique Reynaud
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, UNAM, A.P. 510-3, 62210, Cuernavaca, Mor., Mexico.
| |
Collapse
|
43
|
Kawabe H, Stegmüller J. The role of E3 ubiquitin ligases in synapse function in the healthy and diseased brain. Mol Cell Neurosci 2021; 112:103602. [DOI: 10.1016/j.mcn.2021.103602] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 01/06/2021] [Accepted: 02/02/2021] [Indexed: 02/08/2023] Open
|
44
|
Kim BS, Jang T, Yoo SE, Lee JM, Kim E. Fas-associated factor 1 induces the accumulation of α-synuclein through autophagic suppression in dopaminergic neurons. FASEB J 2021; 35:e21363. [PMID: 33749937 DOI: 10.1096/fj.202001371rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 12/14/2020] [Accepted: 12/28/2020] [Indexed: 12/21/2022]
Abstract
Impairment of protein clearance mechanisms leads to α-synuclein accumulation in dopaminergic neurons, contributing to the pathogenesis of Parkinson's disease (PD). Based on the finding that Fas-associated factor 1 (FAF1), a positive modulator of PD, colocalizes with α-synuclein in PD patient brains, we investigated the existence of pathological interplay between FAF1 and α-synuclein. Monomeric and high-molecular-weight forms of α-synuclein were increased in FAF1-overexpressing SH-SY5Y cells. In particular, α-synuclein turnover was accelerated by genetic depletion of FAF1 in SH-SY5Y cells. Therefore, we questioned whether FAF1 is involved in the α-synuclein clearance process. Autophagy inhibitors, but not proteasome inhibitors, restored concurrent attenuation of α-synuclein expression by FAF1 depletion in SH-SY5Y cells. Moreover, we found alterations in autophagy markers in SH-SY5Y cells caused by FAF1 overexpression, indicating that FAF1 disturbed α-synuclein clearance through the autophagy-lysosome pathway. Indeed, FAF1 activated the mammalian target of rapamycin (mTOR) pathway, subsequently suppressing autophagosome formation. Consistently, α-synuclein-mediated mitochondrial dysfunction was observed in FAF1-overexpressing SH-SY5Y cells. Furthermore, FAF1 overexpression using stereotaxic injection of adeno-associated virus led to α-synuclein accumulation and autophagy dysregulation in the PD model mice. Taken together, our results reveal a novel role for FAF1: that of a negative regulator of autophagic α-synuclein clearance.
Collapse
Affiliation(s)
- Bok-Seok Kim
- Department of Biological Sciences, Chungnam National University, Daejeon, South Korea
| | - Taeik Jang
- Department of Biological Sciences, Chungnam National University, Daejeon, South Korea
| | | | | | - Eunhee Kim
- Department of Biological Sciences, Chungnam National University, Daejeon, South Korea
| |
Collapse
|
45
|
Exploring genetic alterations in circulating tumor DNA from cerebrospinal fluid of pediatric medulloblastoma. Sci Rep 2021; 11:5638. [PMID: 33707557 PMCID: PMC7952732 DOI: 10.1038/s41598-021-85178-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 02/22/2021] [Indexed: 12/27/2022] Open
Abstract
Medulloblastoma (MB) is the most common type of brain malignancy in children. Molecular profiling has become an important component to select patients for therapeutic approaches, allowing for personalized therapy. In this study, we successfully identified detectable levels of tumor-derived cell-free DNA (cfDNA) in cerebrospinal fluid (CSF) samples of patients with MB. Furthermore, cfDNA from CSF can interrogate for tumor-associated molecular clues. MB-associated alterations from CSF, tumor, and post-chemotherapy plasma were compared by deep sequencing on next-generation sequencing platform. Shared alterations exist between CSF and matched tumor tissues. More alternations were detected in circulating tumor DNA from CSF than those in genomic DNA from primary tumor. It was feasible to detect MB-associated mutations in plasma of patients treated with chemotherapy. Collectively, CSF supernatant can be used to monitor genomic alterations, as a superior technique as long as tumor-derived cfDNA can be isolated from CSF successfully.
Collapse
|
46
|
Sian-Hulsmann J, Riederer P. The Nigral Coup in Parkinson's Disease by α-Synuclein and Its Associated Rebels. Cells 2021; 10:598. [PMID: 33803185 PMCID: PMC8000327 DOI: 10.3390/cells10030598] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 12/24/2022] Open
Abstract
The risk of Parkinson's disease increases with age. However, the etiology of the illness remains obscure. It appears highly likely that the neurodegenerative processes involve an array of elements that influence each other. In addition, genetic, endogenous, or exogenous toxins need to be considered as viable partners to the cellular degeneration. There is compelling evidence that indicate the key involvement of modified α-synuclein (Lewy bodies) at the very core of the pathogenesis of the disease. The accumulation of misfolded α-synuclein may be a consequence of some genetic defect or/and a failure of the protein clearance system. Importantly, α-synuclein pathology appears to be a common denominator for many cellular deleterious events such as oxidative stress, mitochondrial dysfunction, dopamine synaptic dysregulation, iron dyshomeostasis, and neuroinflammation. These factors probably employ a common apoptotic/or autophagic route in the final stages to execute cell death. The misfolded α-synuclein inclusions skillfully trigger or navigate these processes and thus amplify the dopamine neuron fatalities. Although the process of neuroinflammation may represent a secondary event, nevertheless, it executes a fundamental role in neurodegeneration. Some viral infections produce parkinsonism and exhibit similar characteristic neuropathological changes such as a modest brain dopamine deficit and α-synuclein pathology. Thus, viral infections may heighten the risk of developing PD. Alternatively, α-synuclein pathology may induce a dysfunctional immune system. Thus, sporadic Parkinson's disease is caused by multifactorial trigger factors and metabolic disturbances, which need to be considered for the development of potential drugs in the disorder.
Collapse
Affiliation(s)
- Jeswinder Sian-Hulsmann
- Department of Medical Physiology, University of Nairobi, P.O. Box 30197, 00100 Nairobi, Kenya
| | - Peter Riederer
- Clinic and Policlinic for Psychiatry, Psychosomatics and Psychotherapy Margarete-Hoeppel-Platz 1, University Hospital Wuerzburg, 97080 Wuerzburg, Germany;
- Department Psychiatry, University of Southern Denmark Odense, J.B. Winslows Vey 18, 5000 Odense, Denmark
| |
Collapse
|
47
|
Liu J, Wang X, Ma R, Li T, Guo G, Ning B, Moran TH, Smith WW. AMPK signaling mediates synphilin-1-induced hyperphagia and obesity in Drosophila. J Cell Sci 2021; 134:jcs247742. [PMID: 33443093 PMCID: PMC7875497 DOI: 10.1242/jcs.247742] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 12/15/2020] [Indexed: 12/31/2022] Open
Abstract
Expression of synphilin-1 in neurons induces hyperphagia and obesity in a Drosophila model. However, the molecular pathways underlying synphilin-1-linked obesity remain unclear. Here, Drosophila models and genetic tools were used to study the synphilin-1-linked pathways in energy balance by combining molecular biology and pharmacological approaches. We found that expression of human synphilin-1 in flies increased AMP-activated kinase (AMPK) phosphorylation at Thr172 compared with that in non-transgenic flies. Knockdown of AMPK reduced AMPK phosphorylation and food intake in non-transgenic flies, and further suppressed synphilin-1-induced AMPK phosphorylation, hyperphagia, fat storage and body weight gain in transgenic flies. Expression of constitutively activated AMPK significantly increased food intake and body weight gain in non-transgenic flies, but it did not alter food intake in the synphilin-1 transgenic flies. In contrast, expression of dominant-negative AMPK reduced food intake in both non-transgenic and synphilin-1 transgenic flies. Treatment with STO-609 also suppressed synphilin-1-induced AMPK phosphorylation, hyperphagia and body weight gain. These results demonstrate that the AMPK signaling pathway plays a critical role in synphilin-1-induced hyperphagia and obesity. These findings provide new insights into the mechanisms of synphilin-1-controlled energy homeostasis.
Collapse
Affiliation(s)
- Jingnan Liu
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA
| | - Xiaobo Wang
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Rui Ma
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA
| | - Tianxia Li
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA
| | - Gongbo Guo
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Bo Ning
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Timothy H Moran
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Wanli W Smith
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
48
|
Interaction between Parkin and α-Synuclein in PARK2-Mediated Parkinson's Disease. Cells 2021; 10:cells10020283. [PMID: 33572534 PMCID: PMC7911026 DOI: 10.3390/cells10020283] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/25/2021] [Accepted: 01/26/2021] [Indexed: 12/12/2022] Open
Abstract
Parkin and α-synuclein are two key proteins involved in the pathophysiology of Parkinson's disease (PD). Neurotoxic alterations of α-synuclein that lead to the formation of toxic oligomers and fibrils contribute to PD through synaptic dysfunction, mitochondrial impairment, defective endoplasmic reticulum and Golgi function, and nuclear dysfunction. In half of the cases, the recessively inherited early-onset PD is caused by loss of function mutations in the PARK2 gene that encodes the E3-ubiquitin ligase, parkin. Parkin is involved in the clearance of misfolded and aggregated proteins by the ubiquitin-proteasome system and regulates mitophagy and mitochondrial biogenesis. PARK2-related PD is generally thought not to be associated with Lewy body formation although it is a neuropathological hallmark of PD. In this review article, we provide an overview of post-mortem neuropathological examinations of PARK2 patients and present the current knowledge of a functional interaction between parkin and α-synuclein in the regulation of protein aggregates including Lewy bodies. Furthermore, we describe prevailing hypotheses about the formation of intracellular micro-aggregates (synuclein inclusions) that might be more likely than Lewy bodies to occur in PARK2-related PD. This information may inform future studies aiming to unveil primary signaling processes involved in PD and related neurodegenerative disorders.
Collapse
|
49
|
Li J, Mo C, Guo Y, Zhang B, Feng X, Si Q, Wu X, Zhao Z, Gong L, He D, Shao J. Roles of peptidyl-prolyl isomerase Pin1 in disease pathogenesis. Theranostics 2021; 11:3348-3358. [PMID: 33537091 PMCID: PMC7847688 DOI: 10.7150/thno.45889] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 12/02/2020] [Indexed: 12/21/2022] Open
Abstract
Pin1 belongs to the peptidyl-prolyl cis-trans isomerases (PPIases) superfamily and catalyzes the cis-trans conversion of proline in target substrates to modulate diverse cellular functions including cell cycle progression, cell motility, and apoptosis. Dysregulation of Pin1 has wide-ranging influences on the fate of cells; therefore, it is closely related to the occurrence and development of various diseases. This review summarizes the current knowledge of Pin1 in disease pathogenesis.
Collapse
Affiliation(s)
- Jingyi Li
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, China
- School of Biological Sciences and Technology, Chengdu Medical College, Chengdu, China
| | - Chunfen Mo
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China
| | - Yifan Guo
- School of Biological Sciences and Technology, Chengdu Medical College, Chengdu, China
| | - Bowen Zhang
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, China
| | - Xiao Feng
- School of Biological Sciences and Technology, Chengdu Medical College, Chengdu, China
| | - Qiuyue Si
- School of Biological Sciences and Technology, Chengdu Medical College, Chengdu, China
| | - Xiaobo Wu
- School of Biological Sciences and Technology, Chengdu Medical College, Chengdu, China
| | - Zhe Zhao
- School of Biological Sciences and Technology, Chengdu Medical College, Chengdu, China
| | - Lixin Gong
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, China
| | - Dan He
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, China
| | - Jichun Shao
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, China
| |
Collapse
|
50
|
Jin M, Li N, Sheng W, Ji X, Liang X, Kong B, Yin P, Li Y, Zhang X, Liu K. Toxicity of different zinc oxide nanomaterials and dose-dependent onset and development of Parkinson's disease-like symptoms induced by zinc oxide nanorods. ENVIRONMENT INTERNATIONAL 2021; 146:106179. [PMID: 33099061 DOI: 10.1016/j.envint.2020.106179] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 09/30/2020] [Accepted: 09/30/2020] [Indexed: 06/11/2023]
Abstract
With the increasing applications in various fields, the release and accumulation of zinc oxide (ZnO) nanomaterials ultimately lead to unexpected consequences to environment and human health. Therefore, toxicity comparison among ZnO nanomaterials with different shape/size and their adverse effects need better characterization. Here, we utilized zebrafish larvae and human neuroblastoma cells SH-SY5Y to compare the toxic effects of ZnO nanoparticles (ZnO NPs), short ZnO nanorods (s-ZnO NRs), and long ZnO NRs (l-ZnO NRs). We found their developmental- and neuro-toxicity levels were similar, where the smaller sizes showed slightly higher toxicity than the larger sizes. The developmental neurotoxicity of l-ZnO NRs (0.1, 1, 10, 50, and 100 μg/mL) was further investigated since they had the lowest toxicity. Our results indicated that l-ZnO NRs induced developmental neurotoxicity with hallmarks linked to Parkinson's disease (PD)-like symptoms at relatively high doses, including the disruption of locomotor activity as well as neurodevelopmental and PD responsive genes expression, and the induction of dopaminergic neuronal loss and apoptosis in zebrafish brain. l-ZnO NRs activated reactive oxygen species production, whose excessive accumulation triggered mitochondrial damage and mitochondrial apoptosis, eventually leading to PD-like symptoms. Collectively, the developmental- and neuro-toxicity of ZnO nanomaterials was identified, in which l-ZnO NRs harbors a remarkably potential risk for the onset and development of PD at relatively high doses, stressing the discretion of safe range in view of nano-ZnO exposure to ecosystem and human beings.
Collapse
Affiliation(s)
- Meng Jin
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Jinan 250103, PR China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Jinan 250103, PR China
| | - Ning Li
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Jinan 250103, PR China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Jinan 250103, PR China
| | - Wenlong Sheng
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Jinan 250103, PR China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Jinan 250103, PR China
| | - Xiuna Ji
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Jinan 250103, PR China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Jinan 250103, PR China
| | - Xiu Liang
- Advanced Materials Institute, Qilu University of Technology (Shandong Academy of Sciences), 19 Keyuan Road, Jinan 250014, PR China.
| | - Biao Kong
- Department of Chemistry, Shanghai Key Lab of Molecular Catalysis and Innovative Materials, iChEM, Fudan University, Shanghai 200433, China
| | - Penggang Yin
- Key Laboratory of Bio-inspired Smart Interfacial Science and Technology of Ministry of Education, School of Chemistry, Beihang University, Beijing 100191, China
| | - Yong Li
- Advanced Materials Institute, Qilu University of Technology (Shandong Academy of Sciences), 19 Keyuan Road, Jinan 250014, PR China
| | - Xingshuang Zhang
- Advanced Materials Institute, Qilu University of Technology (Shandong Academy of Sciences), 19 Keyuan Road, Jinan 250014, PR China
| | - Kechun Liu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Jinan 250103, PR China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Jinan 250103, PR China.
| |
Collapse
|